1
|
Jing S, Zhao L, Zhao L, Gao Y, He T. TRIP13: A promising cancer immunotherapy target. CANCER INNOVATION 2024; 3:e147. [PMID: 39398261 PMCID: PMC11467489 DOI: 10.1002/cai2.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/21/2024] [Accepted: 05/12/2024] [Indexed: 10/15/2024]
Abstract
The tumor microenvironment (TME) facilitates tumor development through intricate intercellular signaling, thereby supporting tumor growth and suppressing the immune response. Thyroid hormone receptor interactor 13 (TRIP13), an AAA+ ATPase, modulates the conformation of client macromolecules, consequently influencing cellular signaling pathways. TRIP13 has been implicated in processes such as proliferation, invasion, migration, and metastasis during tumor progression. Recent studies have revealed that TRIP13 also plays a role in immune response suppression within the TME. Thus, inhibiting these functions of TRIP13 could potentially enhance immune responses and improve the efficacy of immune checkpoint inhibition. This review summarizes the recent research progress of TRIP13 and discusses the potential of targeting TRIP13 to improve immune-based therapies for patients with cancer.
Collapse
Affiliation(s)
- Shengnan Jing
- Institute of Pain Medicine and Special Environmental Medicine, Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| | - Liya Zhao
- Institute of Pain Medicine and Special Environmental Medicine, Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| | - Liwen Zhao
- Institute of Pain Medicine and Special Environmental Medicine, Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| | - Yong‐Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| | - Tianzhen He
- Institute of Pain Medicine and Special Environmental Medicine, Co‐innovation Center of NeuroregenerationNantong UniversityNantongJiangsuChina
| |
Collapse
|
2
|
Grahammer F, Dumoulin B, Gulieva RE, Wu H, Xu Y, Sulaimanov N, Arnold F, Sandner L, Cordts T, Todkar A, Moulin P, Reichardt W, Puelles VG, Kramann R, Freedman BS, Busch H, Boerries M, Walz G, Huber TB. Cyclin-dependent kinase 4 drives cystic kidney disease in the absence of mTORC1 signaling activity. Kidney Int 2024; 106:856-869. [PMID: 39218392 DOI: 10.1016/j.kint.2024.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Progression of cystic kidney disease has been linked to activation of the mTORC1 signaling pathway. Yet the utility of mTORC1 inhibitors to treat patients with polycystic kidney disease remains controversial despite promising preclinical data. To define the cell intrinsic role of mTORC1 for cyst development, the mTORC1 subunit gene Raptor was selectively inactivated in kidney tubular cells lacking cilia due to simultaneous deletion of the kinesin family member gene Kif3A. In contrast to a rapid onset of cyst formation and kidney failure in mice with defective ciliogenesis, both kidney function, cyst formation discerned by magnetic resonance imaging and overall survival were strikingly improved in mice additionally lacking Raptor. However, these mice eventually succumbed to cystic kidney disease despite mTORC1 inactivation. In-depth transcriptome analysis revealed the rapid activation of other growth-promoting signaling pathways, overriding the effects of mTORC1 deletion and identified cyclin-dependent kinase (CDK) 4 as an alternate driver of cyst growth. Additional inhibition of CDK4-dependent signaling by the CDK4/6 inhibitor Palbociclib markedly slowed disease progression in mice and human organoid models of polycystic kidney disease and potentiated the effects of mTORC1 deletion/inhibition. Our findings indicate that cystic kidneys rapidly adopt bypass mechanisms typically observed in drug resistant cancers. Thus, future clinical trials need to consider combinatorial or sequential therapies to improve therapeutic efficacy in patients with cystic kidney disease.
Collapse
Affiliation(s)
- Florian Grahammer
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Bernhard Dumoulin
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ramila E Gulieva
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, Washington, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Hui Wu
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yaoxian Xu
- Institute of Experimental Medicine and Systems Biology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Nurgazy Sulaimanov
- Department of Electrical Engineering and Information Technology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Frederic Arnold
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Lukas Sandner
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Tomke Cordts
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Abhijeet Todkar
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany
| | - Pierre Moulin
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - Wilfried Reichardt
- Department of Diagnostic and Interventional Radiology, Division of Medical Physics, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Rafael Kramann
- Institute of Experimental Medicine and Systems Biology, Medical Faculty, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Benjamin S Freedman
- Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA; Kidney Research Institute, University of Washington School of Medicine, Seattle, Washington, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Plurexa LLC, Seattle, Washington, USA
| | - Hauke Busch
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between Deutsches Krebs Forschungs Zentrum (DKFZ) and Medical Center-University of Freiburg, Heidelberg, Germany
| | - Gerd Walz
- Department of Medicine IV, Medical Center and Faculty of Medicine University of Freiburg, Freiburg, Germany; Signaling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Chen C, Li P, Fan G, Yang E, Jing S, Shi Y, Gong Y, Zhang L, Wang Z. Role of TRIP13 in human cancer development. Mol Biol Rep 2024; 51:1088. [PMID: 39436503 DOI: 10.1007/s11033-024-10012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024]
Abstract
As an AAA + ATPase, thyroid hormone receptor interacting protein 13 (TRIP13) primarily functions in DNA double-strand break repair, chromosome recombination, and cell cycle checkpoint regulation; aberrant expression of TRIP13 can result in chromosomal instability (CIN). According to recent research, TRIP13 is aberrantly expressed in a variety of cancers, and a patient's poor prognosis and tumor stage are strongly correlated with high expression of TRIP13. Tumor cell and subcutaneous xenograft growth can be markedly inhibited by TRIP13 knockdown or TRIP13 inhibitor administration. In the initiation and advancement of human malignancies, TRIP13 seems to function as an oncogene. Based on available data, TRIP13 may function as a biological target and biomarker for cancer. The creation of inhibitors that specifically target TRIP13 may present novel approaches to treating cancer.
Collapse
Affiliation(s)
- Chaohu Chen
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Pan Li
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Guangrui Fan
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Enguang Yang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Suoshi Jing
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yibo Shi
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Yuwen Gong
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Luyang Zhang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, NO.82 Linxia Road, Chengguan District Lanzhou, Lanzhou, Gansu Province, 730030, PR China.
- Gansu Province Clinical Research Center for urinary system disease, Lanzhou, Gansu Province, 730030, PR China.
| |
Collapse
|
4
|
Chen J, Liu Y, Wu X, Zhang Y, Huang W, Han W, Chen G, Xu Q, Chen H, Wu Q, Wang J, Huang J. Identification of a novel splicing variant of thyroid hormone receptor interaction protein 13 (TRIP13) in female infertility characterized by oocyte maturation arrest. J Assist Reprod Genet 2024; 41:2777-2785. [PMID: 39297991 DOI: 10.1007/s10815-024-03219-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/31/2024] [Indexed: 09/21/2024] Open
Abstract
PURPOSE As a cause of primary female infertility, oocyte maturation arrest (OMA) is characterized by failure to obtain mature oocytes due to abnormal meiosis. We aimed to identify pathogenic variants in two sisters with OMA phenotype from a non-consanguineous family. METHODS Whole-exome sequencing and Sanger sequencing were conducted to identify and validate the disease-causing gene variant. Additionally, we performed a minigene assay, quantitative reverse transcription PCR, and Western blotting to assess the effects of the variant. RESULTS We identified a novel homozygous splicing variant (c.1021-11T>C) in TRIP13, which followed a recessive inheritance pattern. Minigene assay showed that the variant could disrupt the integrity of TRIP13 mRNA, as evidenced by the production of an alternative transcript with intron10 intermediate retention of 79 bp. Compared to normal controls, the expression of TRIP13 mRNA and abundance of TRIP13 protein were also significantly decreased in Epstein-Barr virus-immortalized lymphoblastoid cells derived from affected individuals. CONCLUSION Our findings confirm the contribution of genetic factors to OMA and expand the mutation spectrum of TRIP13 in female infertility.
Collapse
Affiliation(s)
- Jia Chen
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Yuxin Liu
- Department of Clinical Medicine, Nanchang University School of Queen Mary, Nanchang, China
| | - Xingwu Wu
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Yiwei Zhang
- Department of Clinical Medicine, Nanchang University School of Queen Mary, Nanchang, China
| | - Wen Huang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Wenbo Han
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Ge Chen
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Qiang Xu
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Houyang Chen
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Qiongfang Wu
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China
| | - Jiawei Wang
- Division of Life Sciences and Medicine, Reproductive and Genetic Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China.
| | - Jialyu Huang
- Center for Reproductive Medicine, Jiangxi Key Laboratory of Reproductive Health, Jiangxi Branch of National Clinical Research Center for Obstetrics and Gynecology, Jiangxi Maternal and Child Health Hospital, Nanchang Medical College, Nanchang, China.
| |
Collapse
|
5
|
Jacob Bunu S, Cai H, Wu L, Zhang H, Zhou Z, Xu Z, Shi J, Zhu W. TRIP13 - a potential drug target in cancer pharmacotherapy. Bioorg Chem 2024; 151:107650. [PMID: 39042962 DOI: 10.1016/j.bioorg.2024.107650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/25/2024]
Abstract
ATPases Associated with Diverse Cellular Activity (AAA+ATPases) are important enzymatic functional proteins in human cells. Thyroid Hormone Receptor Interacting Protein-13 (TRIP13) is a member of this protein superfamily, that partly regulates DNA repair pathways and spindle assembly checkpoints during mitosis. TRIP13 is reported as an oncogene involving multiple pathways in many human malignancies, including multiple myeloma, brain tumors, etc. The structure of TRIP13 reveals the mechanisms for ATP binding and how TRIP13 recognizes the Mitotic Arrest Deficiency-2 (MAD2) protein, with p31comet acting as an adapter protein. DCZ0415, TI17, DCZ5417, and DCZ5418 are the reported small-molecule inhibitors of TRIP13, which have been demonstrated to inhibit TRIP13's biological functions significantly and effective in suppressing various types of malignant cells, indicating that TRIP13 is a significant anticancer drug target. Currently, no systematic reviews are cutting across the functions, structure, and novel inhibitors of TRIP13. This review provides a comprehensive overview of TRIP13's biological functions, its roles in eighteen different cancers, four small molecule inhibitors, different underlying molecular mechanisms, and its functionality as a potential anticancer drug target.
Collapse
Affiliation(s)
- Samuel Jacob Bunu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Haiyan Cai
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Leyun Wu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Hui Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhaoyin Zhou
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
6
|
Ding Y, Huang K, Sun C, Liu Z, Zhu J, Jiao X, Liao Y, Feng X, Guo J, Zhu C, Zhai Z, Xiong S. A Bruton tyrosine kinase inhibitor-resistance gene signature predicts prognosis and identifies TRIP13 as a potential therapeutic target in diffuse large B-cell lymphoma. Sci Rep 2024; 14:21184. [PMID: 39261532 PMCID: PMC11391086 DOI: 10.1038/s41598-024-72121-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024] Open
Abstract
Bruton tyrosine kinase inhibitor (BTKi) combined with rituximab-based chemotherapy benefits diffuse large B-cell lymphoma (DLBCL) patients. However, drug resistance is the major cause of relapse and death of DLBCL. In this study, we conducted a comprehensive analysis BTKi-resistance related genes (BRRGs) and established a 10-gene (CARD16, TRIP13, PSRC1, CASP1, PLBD1, CARD6, CAPG, CACNA1A, CDH15, and NDUFA4) signature for early identifying high-risk DLBCL patients. The resistance scores based on the BRRGs signature were associated with prognosis. Furthermore, we developed a nomogram incorporating the BRRGs signature, which demonstrated excellent performance in predicting the prognosis of DLBCL patients. Notably, tumor immune microenvironment, biological pathways, and chemotherapy sensitivity were different between high- and low-resistance score groups. Additionally, we identified TRIP13 as a key gene in our model. TRIP13 was found to be overexpressed in DLBCL and BTKi-resistant DLBCL cell lines, knocking down TRIP13 suppresses cell proliferation, promotes cell apoptosis, and enhances the apoptosis effect of BTKi on DLBCL cells by regulating the Wnt/β-catenin pathway. In conclusion, our study presents a novel BRRGs signature that could serve as a promising prognostic marker in DLBCL, and TRIP13 might be a potential therapeutic target for resistant DLBCL.
Collapse
Affiliation(s)
- Yangyang Ding
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Keke Huang
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Cheng Sun
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Zelin Liu
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jinli Zhu
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xunyi Jiao
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Ya Liao
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xiangjiang Feng
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jingjing Guo
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Chunhua Zhu
- Air Force Health Care Center for Special Services, Hangzhou, Zhejiang, People's Republic of China
| | - Zhimin Zhai
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Shudao Xiong
- Department of Hematology/Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.
| |
Collapse
|
7
|
Li X, Zhang H, Dong S, Gao X, Sun H, Zhou Z, Hu K, Guo S, Zhang Q, Guo Z, Jacob Bunu S, Zhu J, Li B, Zhang Y, Shen J, Akber Aisa H, Xu Z, Cai H, Shi J, Zhu W. Design, synthesis, and biological evaluation of novel 1-amido-2-one-4-thio-deoxypyranose as potential antitumor agents for multiple myeloma. Bioorg Med Chem 2024; 111:117843. [PMID: 39083980 DOI: 10.1016/j.bmc.2024.117843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
This study reported the design and synthesis of novel 1-amido-2-one-4-thio-deoxypyranose as inhibitors of potential drug target TRIP13 for developing new mechanism-based therapeutic agents in the treatment of multiple myeloma (MM). In comparison with the positive control DCZ0415, the most active compounds C16, C18, C20 and C32 exhibited strong anti-proliferative activity against human MM cell lines (ARP-1 and NCI-H929) with IC50 values of 1 ∼ 2 μM. While the surface plasmon resonance (SPR) and ATPase activity assays demonstrated that the representative compound C20 is a potent inhibitor of TRIP13, C20 also showed good antitumor activity in vivo on BALB/c nude mice xenografted with MM tumor cells. An initial structure-activity study showed that the carbonyl group is crucial for anticancer activity. Overall, this study provided novel 1-amido-2-one-4-thio-deoxypyranoses, which are entirely different from previously reported potent inhibitor structures of TRIP13, and thus would aid the development of carbohydrate-based novel agents in MM pharmacotherapy.
Collapse
Affiliation(s)
- Xiaomei Li
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, CAS Laboratory of Chemistry of Plant Resources in Arid Regions, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, Xinjiang, China; State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Hui Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Sanfeng Dong
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Xuejie Gao
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Haiguo Sun
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Zhaoyin Zhou
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Ke Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Shushan Guo
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Qikai Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhufeng Guo
- School of Resources and Environmental Engineering, Shanghai Polytechnic University, No. 2360 Jinhai Road, Shanghai 201209, China
| | - Samuel Jacob Bunu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Jianming Zhu
- School of Resources and Environmental Engineering, Shanghai Polytechnic University, No. 2360 Jinhai Road, Shanghai 201209, China
| | - Bo Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Yong Zhang
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Jingshan Shen
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Haji Akber Aisa
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, CAS Laboratory of Chemistry of Plant Resources in Arid Regions, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, Xinjiang, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China.
| | - Haiyan Cai
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Weiliang Zhu
- State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, CAS Laboratory of Chemistry of Plant Resources in Arid Regions, Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi, Xinjiang, China; State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing 100049, China.
| |
Collapse
|
8
|
Zhang G, Yang R, Wang B, Yan Q, Zhao P, Zhang J, Su W, Yang L, Cui H. TRIP13 regulates progression of gastric cancer through stabilising the expression of DDX21. Cell Death Dis 2024; 15:622. [PMID: 39187490 PMCID: PMC11347623 DOI: 10.1038/s41419-024-07012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 08/28/2024]
Abstract
GC (Gastric cancer) is one of the most common malignant tumours, with over 95% of gastric cancer patients being adenocarcinoma and most gastric cancer patients having no apparent symptoms in the early stages. Finding biomarkers for early screening of gastric cancer and exploring new targets for gastric cancer treatment are urgent problems to be solved in the treatment of gastric cancer, with significant clinical outcomes for the survival rate of gastric cancer patients. The AAA+ family ATPase thyroid hormone receptor-interacting protein 13 (TRIP13) has been reported to play an essential role in developing various tumours. However, the biological function and molecular mechanism of TRIP13 in gastric cancer remain unclear. This study confirms that TRIP13 is highly expressed in gastric cancer tissue samples and that TRIP13 participates in the proliferation, migration, invasion in vitro, and tumourigenesis and metastasis in vivo of gastric cancer cells. Mechanistically, this study confirms that TRIP13 directly interacts with DDX21 and stabilises its expression by restraining its ubiquitination degradation, thereby promoting gastric cancer progression. Additionally, histone deacetylase 1 (HDAC1) is an upstream factor of TRIP13, which could target the TRIP13 promoter region to promote the proliferation, migration, and invasion of gastric cancer cells. These results indicate that TRIP13 serve is a promising biomarker for the treating of gastric cancer patients, and the HDAC1-TRIP13/DDX21 axis might provide a solid theoretical basis for clinical treatment of gastric cancer patients.
Collapse
Affiliation(s)
- Guanghui Zhang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China.
| | - Rui Yang
- Biomedical Laboratory, School of Medicine, Liaocheng University, Liaocheng, China
| | - Baiyan Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Qiujin Yan
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peiyuan Zhao
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiaming Zhang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Weiyu Su
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lianhe Yang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China.
| |
Collapse
|
9
|
Swanson M, Yun J, Collier DM, Challagundla L, Dogan M, Kuscu C, Garrett MR, Regner KR, Chung JH, Park F. Removal of the catalytic subunit of DNA-protein kinase in the proximal tubules promotes DNA and tubular damage during kidney injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609216. [PMID: 39229063 PMCID: PMC11370575 DOI: 10.1101/2024.08.22.609216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Tubular epithelial cell damage can be repaired through a series of complex signaling pathways. An early event in many forms of tubular damage is the observation of DNA damage, which can be repaired by specific pathways depending upon the type of genomic alteration.. In this study, we report that the catalytic subunit of DNA protein kinase (DNA-PKcs), a central DNA repair enzyme involved in sensing DNA damage and performing double stranded DNA break repair, plays an important role in the extent of tubular epithelial cell damage following exposure to injurious acute and chronic stimuli. Selective loss of DNA-PKcs in the proximal tubules led to increased markers of kidney dysfunction, DNA damage, and tubular epithelial cell injury in multiple models of acute kidney injury, specifically bilateral renal ischemia-reperfusion injury and single dose of cisplatin (15 mg/kg IP). In contrast, in a mouse model of kidney fibrosis and chronic kidney disease (UUO),the protective effects of DNA-PKcs was not as obvious histologically from the tissue sections. In the absence of proximal tubular DNA-PKcs, there was reduced levels of fibrotic markers, α-SMA and fibronectin, which suggests that there may be a biphasic role of DNA-PKcs depending upon the conditions exerted upon the kidney. In conclusion, this study demonstrates that the catalytic subunit of DNA-PKcs plays a context-dependent role in the kidney to reduce DNA damage during exposure to various types of acute, but not chronic forms of injurious stimuli.
Collapse
|
10
|
Zin I, China A, Khan K, Nag JK, Vasu K, Deshpande GM, Ghosh PK, Khan D, Ramachandiran I, Ganguly S, Tamagno I, Willard B, Gogonea V, Fox PL. AKT-dependent nuclear localization of EPRS1 activates PARP1 in breast cancer cells. Proc Natl Acad Sci U S A 2024; 121:e2303642121. [PMID: 39012819 PMCID: PMC11287164 DOI: 10.1073/pnas.2303642121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/05/2024] [Indexed: 07/18/2024] Open
Abstract
Glutamyl-prolyl-tRNA synthetase (EPRS1) is a bifunctional aminoacyl-tRNA-synthetase (aaRS) essential for decoding the genetic code. EPRS1 resides, with seven other aaRSs and three noncatalytic proteins, in the cytoplasmic multi-tRNA synthetase complex (MSC). Multiple MSC-resident aaRSs, including EPRS1, exhibit stimulus-dependent release from the MSC to perform noncanonical activities distinct from their primary function in protein synthesis. Here, we show EPRS1 is present in both cytoplasm and nucleus of breast cancer cells with constitutively low phosphatase and tensin homolog (PTEN) expression. EPRS1 is primarily cytosolic in PTEN-expressing cells, but chemical or genetic inhibition of PTEN, or chemical or stress-mediated activation of its target, AKT, induces EPRS1 nuclear localization. Likewise, preferential nuclear localization of EPRS1 was observed in invasive ductal carcinoma that were also P-Ser473-AKT+. EPRS1 nuclear transport requires a nuclear localization signal (NLS) within the linker region that joins the catalytic glutamyl-tRNA synthetase and prolyl-tRNA synthetase domains. Nuclear EPRS1 interacts with poly(ADP-ribose) polymerase 1 (PARP1), a DNA-damage sensor that directs poly(ADP-ribosyl)ation (PARylation) of proteins. EPRS1 is a critical regulator of PARP1 activity as shown by markedly reduced ADP-ribosylation in EPRS1 knockdown cells. Moreover, EPRS1 and PARP1 knockdown comparably alter the expression of multiple tumor-related genes, inhibit DNA-damage repair, reduce tumor cell survival, and diminish tumor sphere formation by breast cancer cells. EPRS1-mediated regulation of PARP1 activity provides a mechanistic link between PTEN loss in breast cancer cells, PARP1 activation, and cell survival and tumor growth. Targeting the noncanonical activity of EPRS1, without inhibiting canonical tRNA ligase activity, provides a therapeutic approach potentially supplementing existing PARP1 inhibitors.
Collapse
Affiliation(s)
- Isaac Zin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
- Department of Chemistry, Cleveland State University, Cleveland, OH44115
| | - Arnab China
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Krishnendu Khan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
- Department of Life Sciences, School of Science, Gandhi Institute of Technology and Management, Bengaluru562163, India
| | - Jeetendra K. Nag
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Kommireddy Vasu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | | | - Prabar K. Ghosh
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Debjit Khan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Iyappan Ramachandiran
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Shinjini Ganguly
- Translational Hematology and Oncology Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Ilaria Tamagno
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
| | - Belinda Willard
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| | - Valentin Gogonea
- Department of Chemistry, Cleveland State University, Cleveland, OH44115
| | - Paul L. Fox
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH44195
- Department of Chemistry, Cleveland State University, Cleveland, OH44115
| |
Collapse
|
11
|
Liu L, Liao B, Fan R, Liu Y, Li A, Liu L, Li Y, Li J. TRIP13 Plays an Important Role in the Sensitivity of Leukemia Cell Response to Sulforaphane Therapy. ACS OMEGA 2024; 9:26628-26640. [PMID: 38911763 PMCID: PMC11191565 DOI: 10.1021/acsomega.4c03450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
Sulforaphane is one of the most characterized isothiocyanate compounds in cruciferous vegetables and shows anticancer effects, especially antileukemia properties. However, the molecular mechanism of the growth inhibition effect of sulforaphane in acute myeloid leukemia (AML) has not been fully explored. In the present study, a proteomic analysis was performed on the AML cell line U937 responding to sulforaphane treatment to identify novel and efficient therapeutic targets of sulforaphane on AML cells. Key driver analysis was run on the leukemia network, and TRIP13 was identified as a key regulatory factor in sulforaphane-induced growth inhibition in U937 cells. Pretreatment with DCZ0415, an inhibitor of TRIP13, could significantly attenuate sulforaphane-induced cell apoptosis and cell cycle arrest in vitro through the PI3K/Akt/mTOR signaling pathway. In addition, the inhibitory effect of sulforaphane on the tumor volume could also be obviously attenuated by the pretreatment of DCZ0415 in vivo. These results indicate that TRIP13 plays an important role in the sensitivity of leukemia cell response to sulforaphane treatment, and these findings expand the understanding of the mechanism of the antileukemic effect of sulforaphane and provide a new target for the treatment of AML.
Collapse
Affiliation(s)
- Lei Liu
- Medical
Research Center, The Third People’s Hospital of Chengdu (Affiliated
Hospital of Southwest Jiaotong University), College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Baixue Liao
- College
of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Ruiling Fan
- School
of Pharmacy, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Yanxia Liu
- College
of Pharmacy, Third Military Medical University
(Army Medical University), Chongqing 400038, China
| | - Aoshuang Li
- College
of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Lüye Liu
- Medical
Research Center, The Third People’s Hospital of Chengdu (Affiliated
Hospital of Southwest Jiaotong University), College of Medicine, Southwest Jiaotong University, Chengdu 610031, Sichuan, China
| | - Yan Li
- Department
of General Surgery, The 77th Army Hospital, Leshan 614000, Sichuan, China
| | - Jing Li
- Department
of Pharmacological Research Lab, The Beibei
Affiliated Hospital of Chongqing Medical University, The Ninth People’s
Hospital of Chongqing, Chongqing 400799, China
| |
Collapse
|
12
|
Chen R, Zhu S, Zhao R, Liu W, Jin L, Ren X, He H. Targeting ferroptosis as a potential strategy to overcome the resistance of cisplatin in oral squamous cell carcinoma. Front Pharmacol 2024; 15:1402514. [PMID: 38711989 PMCID: PMC11071065 DOI: 10.3389/fphar.2024.1402514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 03/29/2024] [Indexed: 05/08/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a crucial public health problem, accounting for approximately 2% of all cancers globally and 90% of oral malignancies over the world. Unfortunately, despite the achievements in surgery, radiotherapy, and chemotherapy techniques over the past decades, OSCC patients still low 5-year survival rate. Cisplatin, a platinum-containing drug, serves as one of the first-line chemotherapeutic agents of OSCC. However, the resistance to cisplatin significantly limits the clinical practice and is a crucial factor in tumor recurrence and metastasis after conventional treatments. Ferroptosis is an iron-based form of cell death, which is initiated by the intracellular accumulation of lipid peroxidation and reactive oxygen species (ROS). Interestingly, cisplatin-resistant OSCC cells exhibit lower level of ROS and lipid peroxidation compared to sensitive cells. The reduced ferroptosis in cisplatin resistance cells indicates the potential relationship between cisplatin resistance and ferroptosis, which is proved by recent studies showing that in colorectal cancer cells. However, the modulation pathway of ferroptosis reversing cisplatin resistance in OSCC cells still remains unclear. This article aims to concisely summarize the molecular mechanisms and evaluate the relationship between ferroptosis and cisplatin resistance OSCC cells, thereby providing novel strategies for overcoming cisplatin resistance and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Rongkun Chen
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Shuyu Zhu
- Department of Oral Implantology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Ruoyu Zhao
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Wang Liu
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Luxin Jin
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Xiaobin Ren
- Yunnan Key Laboratory of Stomatology, School of Stomatology, Kunming Medical University, Kunming, China
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| | - Hongbing He
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming Medical University, Kunming, China
| |
Collapse
|
13
|
Chen G, Gao X, Jia X, Wang Y, Xu L, Yu D, Chang S, Deng H, Hu K, Wang G, Li B, Xu Z, Lu Y, Wang H, Zhang T, Song D, Yang G, Wu X, Zhu H, Zhu W, Shi J. Ribosomal protein S3 mediates drug resistance of proteasome inhibitor: potential therapeutic application in multiple myeloma. Haematologica 2024; 109:1206-1219. [PMID: 37767568 PMCID: PMC10985453 DOI: 10.3324/haematol.2023.282789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Multiple myeloma (MM) remains incurable due to drug resistance. Ribosomal protein S3 (RPS3) has been identified as a non-Rel subunit of NF-κB. However, the detailed biological roles of RPS3 remain unclear. Here, we report for the first time that RPS3 is necessary for MM survival and drug resistance. RPS3 was highly expressed in MM, and knockout of RPS3 in MM inhibited cell growth and induced cell apoptosis both in vitro and in vivo. Overexpression of RPS3 mediated the proteasome inhibitor resistance of MM and shortened the survival of MM tumor-bearing animals. Moreover, our present study found an interaction between RPS3 and the thyroid hormone receptor interactor 13 (TRIP13), an oncogene related to MM tumorigenesis and drug resistance. We demonstrated that the phosphorylation of RPS3 was mediated by TRIP13 via PKCδ, which played an important role in activating the canonical NF-κB signaling and inducing cell survival and drug resistance in MM. Notably, the inhibition of NF-κB signaling by the small-molecule inhibitor targeting TRIP13, DCZ0415, was capable of triggering synergistic cytotoxicity when combined with bortezomib in drug-resistant MM. This study identifies RPS3 as a novel biomarker and therapeutic target in MM.
Collapse
Affiliation(s)
- Gege Chen
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Xuejie Gao
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Xinyan Jia
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Yingcong Wang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Li Xu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Dandan Yu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Shuaikang Chang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Hui Deng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Ke Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Guanli Wang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120
| | - Bo Li
- CAS Key Laboratory of Receptor Research; State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research; State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203
| | - Yumeng Lu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Huaping Wang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Ting Zhang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Dongliang Song
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Guang Yang
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Xiaosong Wu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Huabin Zhu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research; State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203.
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120.
| |
Collapse
|
14
|
GE S, GU R, YANG X, XU C, WANG S, ZHU G. [TRIP13 Enhances Radioresistance of Lung Adenocarcinoma Cells
through the Homologous Recombination Pathway]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:1-12. [PMID: 38296621 PMCID: PMC10895292 DOI: 10.3779/j.issn.1009-3419.2023.106.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Radiation therapy is one of the most common treatments for non-small cell lung cancer (NSCLC). However, the insensitivity of some tumor cells to radiation is one of the major reasons for the poor efficacy of radiotherapy and the poor prognosis of patients, and exploring the underlying mechanisms behind radioresistance is the key to solving this clinical challenge. This study aimed to identify the molecules associated with radioresistance in lung adenocarcinoma (LUAD), identified thyroid hormone receptor interactor 13 (TRIP13) as the main target initially, and explored whether TRIP13 is related to radioresistance in LUAD and the specific mechanism, with the aim of providing theoretical basis and potential targets for the combination therapy of LUAD patients receiving radiotherapy in the clinic. METHODS Three datasets, GSE18842, GSE19188 and GSE33532, were selected from the Gene Expression Omnibus (GEO) database and screened for differentially expressed genes (|log FC|>1.5, P<0.05) in each of the three datasets using the R 4.1.3 software, and then Venn diagram was used to find out the differentially expressed genes common to the three datasets. The screened differential genes were then subjected to protein-protein interaction (PPI) analysis and module analysis with the help of STRING online tool and Cytoscape software, and survival prognosis analysis was performed for each gene with the help of Kaplan-Meier Plotter database, and the TRIP13 gene was identified as the main molecule for subsequent studies. Subsequently, the human LUAD cell line H292 was irradiated with multiple X-rays using a sub-lethal dose irradiation method to construct a radioresistant cell line, H292DR. The radioresistance of H292DR cells was verified using cell counting kit-8 (CCK-8) assay and clone formation assay. The expression levels of TRIP13 in H292 and H292DR cells were measured by Western blot. Small interfering RNA (siRNA) was used to silence the expression of TRIP13 in H292DR cells and Western blot assay was performed. The clone formation ability and migration ability of H292DR cells were observed after TRIP13 silencing, followed by the detection of changes in the expression levels of proteins closely related to homologous recombination, such as ataxia telangiectasia mutated (ATM) protein. RESULTS Screening of multiple GEO datasets, validation of external datasets and survival analysis revealed that TRIP13 was highly expressed in LUAD and was associated with poor prognosis in LUAD patients who had received radiation therapy. And the results of gene set enrichment analysis (GSEA) of TRIP13 suggested that TRIP13 might be closely associated with LUAD radioresistance by promoting homologous recombination repair after radiation therapy. Experimentally, TRIP13 expression was found to be upregulated in H292DR, and silencing of TRIP13 was able to increase the sensitivity of H292DR cells to radiation. CONCLUSIONS TRIP13 is associated with poor prognosis in LUAD patients treated with radiation, possibly by promoting a homologous recombination repair pathway to mediate resistance of LUAD cells to radiation.
Collapse
|
15
|
Liu W, Bruggeman JW, Lei Q, van Pelt AMM, Koster J, Hamer G. Germline specific genes increase DNA double-strand break repair and radioresistance in lung adenocarcinoma cells. Cell Death Dis 2024; 15:38. [PMID: 38216586 PMCID: PMC10786935 DOI: 10.1038/s41419-024-06433-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 12/18/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024]
Abstract
In principle, germline cells possess the capability to transmit a nearly unaltered set of genetic material to infinite future generations, whereas somatic cells are limited by strict growth constraints necessary to assure an organism's physical structure and eventual mortality. As the potential to replicate indefinitely is a key feature of cancer, we hypothesized that the activation of a "germline program" in somatic cells can contribute to oncogenesis. Our group recently described over one thousand germline specific genes that can be ectopically expressed in cancer, yet how germline specific processes contribute to the malignant properties of cancer is poorly understood. We here show that the expression of germ cell/cancer (GC) genes correlates with malignancy in lung adenocarcinoma (LUAD). We found that LUAD cells expressing more GC genes can repair DNA double strand breaks more rapidly, show higher rates of proliferation and are more resistant to ionizing radiation, compared to LUAD cells that express fewer GC genes. In particular, we identified the HORMA domain protein regulator TRIP13 to be predominantly responsible for this malignant phenotype, and that TRIP13 inhibition or expression levels affect the response to ionizing radiation and subsequent DNA repair. Our results demonstrate that GC genes are viable targets in oncology, as they induce increased radiation resistance and increased propagation in cancer cells. Because their expression is normally restricted to germline cells, we anticipate that GC gene directed therapeutic options will effectively target cancer, with limited side effects besides (temporary) infertility.
Collapse
Affiliation(s)
- Wenqing Liu
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Jan Willem Bruggeman
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Qijing Lei
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Ans M M van Pelt
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Jan Koster
- Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Geert Hamer
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Afaq F, Agarwal S, Bajpai P, Diffalha SA, Kim HG, Peter S, Khushman M, Chauhan SC, Mukherjee P, Varambally S, Manne U. Targeting of oncogenic AAA-ATPase TRIP13 reduces progression of pancreatic ductal adenocarcinoma. Neoplasia 2024; 47:100951. [PMID: 38039923 PMCID: PMC10716004 DOI: 10.1016/j.neo.2023.100951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/21/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
Thyroid hormone receptor-interacting protein 13 (TRIP13) is involved in cancer progression, but its role in pancreatic ductal adenocarcinoma (PDAC) is unknown. Thus, we assessed the expression, functional role, and mechanism of action of TRIP13 in PDAC. We further examined the efficacy of TRIP13 inhibitor, DCZ0415, alone or in combination with gemcitabine on malignant phenotypes, tumor progression, and immune response. We found that TRIP13 was overexpressed in human PDACs relative to corresponding normal pancreatic tissues. TRIP13 knockdown or treatment of PDAC cells with DCZ0415 reduced proliferation and colony formation, and induced G2/M cell cycle arrest and apoptosis. Additionally, TRIP13 knockdown or targeting with DCZ0415 reduced the migration and invasion of PDAC cells by increasing E-cadherin and decreasing N-cadherin and vimentin. Pharmacologic targeting or silencing of TRIP13 also resulted in reduce expression of FGFR4 and STAT3 phosphorylation, and downregulation of the Wnt/β-catenin pathway. In immunocompromised mouse models of PDAC, knockdown of TRIP13 or treatment with DCZ0415 reduced tumor growth and metastasis. In an immunocompetent syngeneic PDAC model, DCZ0415 treatment enhanced the immune response by lowering expression of PD1/PDL1, increasing granzyme B/perforin expression, and facilitating infiltration of CD3/CD4 T-cells. Further, DCZ0415 potentiated the anti-metastatic and anti-tumorigenic activities of gemcitabine by reducing proliferation and angiogenesis and by inducing apoptosis and the immune response. These preclinical findings show that TRIP13 is involved in PDAC progression and targeting of TRIP13 augments the anticancer effect of gemcitabine.
Collapse
Affiliation(s)
- Farrukh Afaq
- Department of Pathology, University of Alabama at Birmingham, USA
| | - Sumit Agarwal
- Department of Pathology, University of Alabama at Birmingham, USA
| | - Prachi Bajpai
- Department of Pathology, University of Alabama at Birmingham, USA
| | - Sameer Al Diffalha
- Department of Pathology, University of Alabama at Birmingham, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, USA
| | - Hyung-Gyoon Kim
- Department of Pathology, University of Alabama at Birmingham, USA
| | - Shajan Peter
- Department of Medicine, Division of Gastroenterology, University of Alabama at Birmingham, USA
| | - Moh'd Khushman
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, USA
| | - Priyabrata Mukherjee
- Department of Pathology, the University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, USA
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, USA.
| |
Collapse
|
17
|
Chang S, Xiao W, Xie Y, Xu Z, Li B, Wang G, Hu K, Zhang Y, Zhou J, Song D, Zhu H, Wu X, Lu Y, Shi J, Zhu W. TI17, a novel compound, exerts anti-MM activity by impairing Trip13 function of DSBs repair and enhancing DNA damage. Cancer Med 2023; 12:21321-21334. [PMID: 37942576 PMCID: PMC10726904 DOI: 10.1002/cam4.6706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/08/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Thyroid hormone receptor interacting protein 13 (Trip13) is an AAA-ATPase that regulates the assembly or disassembly protein complexes and mediates Double-strand breaks (DSBs) repair. Overexpression of Trip13 has been detected in many cancers and is associated with myeloma progression, disease relapse and poor prognosis inmultiple myeloma (MM). METHODS We have identified a small molecular, TI17, through a parallel compound-centric approach, which specifically targets Trip13. To identify whether TI17 targeted Trip13, pull-down and nuclear magnetic resonance spectroscopy (NMR) assays were performed. Cell counting kit-8, clone formation, apoptosis and cell cycle assays were applied to investigate the effects of TI17. We also utilized a mouse model to investigate the effects of TI17 in vivo. RESULTS TI17 effectively inhibited the proliferation of MM cells, and induced the cycle arrest and apoptosis of MM cells. Furthermore, treatment with TI17 abrogates tumor growth and has no apparent side effects in mouse xenograft models. TI17 specifically impaired Trip13 function of DSBs repair and enhanced DNA damage responses in MM. Combining with melphalan or HDAC inhibitor panobinostat triggers synergistic anti-MM effect. CONCLUSIONS Our study suggests that TI17 could be acted as a specific inhibitor of Trip13 and supports a preclinical proof of concept for therapeutic targeting of Trip13 in MM.
Collapse
Affiliation(s)
- Shuaikang Chang
- Department of Hematology, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Wenqin Xiao
- Department of Gastroenterology, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yongsheng Xie
- Department of Hematology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Bo Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Guanli Wang
- Department of Hematology, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Ke Hu
- Department of Hematology, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yong Zhang
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| | - Jinfeng Zhou
- Department of Hematology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Dongliang Song
- Department of Hematology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Huabin Zhu
- Department of Hematology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Xiaosong Wu
- Department of Hematology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Yumeng Lu
- Department of Hematology, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Jumei Shi
- Department of Hematology, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
| |
Collapse
|
18
|
Zhao L, Ye S, Jing S, Gao YJ, He T. Targeting TRIP13 for overcoming anticancer drug resistance (Review). Oncol Rep 2023; 50:202. [PMID: 37800638 PMCID: PMC10565899 DOI: 10.3892/or.2023.8639] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023] Open
Abstract
Cancer is one of the greatest dangers to human wellbeing and survival. A key barrier to effective cancer therapy is development of resistance to anti‑cancer medications. In cancer cells, the AAA+ ATPase family member thyroid hormone receptor interactor 13 (TRIP13) is key in promoting treatment resistance. Nonetheless, knowledge of the molecular processes underlying TRIP13‑based resistance to anticancer therapies is lacking. The present study evaluated the function of TRIP13 expression in anticancer drug resistance and potential methods to overcome this resistance. Additionally, the underlying mechanisms by which TRIP13 promotes resistance to anticancer drugs were explored, including induction of mitotic checkpoint complex surveillance system malfunction, promotion of DNA repair, the enhancement of autophagy and the prevention of immunological clearance. The effects of combination treatment, which include a TRIP13 inhibitor in addition to other inhibitors, were discussed. The present study evaluated the literature on TRIP13 as a possible target and its association with anticancer drug resistance, which may facilitate improvements in current anticancer therapeutic options.
Collapse
Affiliation(s)
- Liwen Zhao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Siyu Ye
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Shengnan Jing
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| | - Tianzhen He
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226019, P.R. China
| |
Collapse
|
19
|
Xue J, Wu H, Shi Y, Li Z. TRIP13 overexpression in hepatocellular carcinoma: implications for poor prognosis and immune cell infiltration. Discov Oncol 2023; 14:176. [PMID: 37740123 PMCID: PMC10516817 DOI: 10.1007/s12672-023-00792-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
PURPOSE The overexpression of TRIP13 has been observed in many types of cancer and has been identified as an oncogene. However, its role in hepatocellular carcinoma (HCC) has not been extensively studied. This study aimed to investigate the expression of TRIP13 in HCC and its impact on immune cell infiltration and prognosis. METHODS We analyzed TCGA and GSE62232 datasets to assess TRIP13 expression in HCC. Kaplan-Meier and subgroup analysis were performed to examine the correlation between TRIP13 expression and HCC. Univariate and Cox regression analysis were conducted to determine the predictive value of TRIP13 in assessing patient outcomes. A nomogram was developed using TRIP13 mRNA expression to predict HCC prognosis. TRIP13 expression was validated using immunohistochemistry in our patient cohort. Survival and subgroup analyses were conducted to investigate the role of TRIP13 in HCC prognosis. RESULTS The results indicated that TRIP13 upregulation in HCC was a strong independent predictor of poor outcome, as determined by Kaplan-Meier and Cox regression analyses. A high AUC value of 0.982 from ROC curves suggested that TRIP13 upregulation could serve as a reliable diagnostic indicator for HCC. The immunohistochemical validation of TRIP13 expression in the patient cohort confirmed its prognostic significance, and high TRIP13 expression was found to be associated with increased infiltration of Th2 cells and decreased infiltration of neutrophils, Th17 cells, and dendritic cells. CONCLUSION These findings suggest that TRIP13 could be a potential prognostic biomarker for HCC.
Collapse
Affiliation(s)
- Jiapeng Xue
- Department of General Surgery, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Hongfen Wu
- Department of Gastroenterology, West China (Sanya) Hospital, Sichuan University, Sanya, China
| | - Yun Shi
- Department of General Surgery, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Zhi Li
- Department of General Surgery, Hubei Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Hubei Provincial Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
20
|
Hu L, Shi J, Shen D, Zhai X, Liang D, Wang J, Xie C, Xia Z, Cui J, Liu F, Du S, Meng S, Piao H. Osimertinib induces paraptosis and TRIP13 confers resistance in glioblastoma cells. Cell Death Discov 2023; 9:333. [PMID: 37669963 PMCID: PMC10480197 DOI: 10.1038/s41420-023-01632-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023] Open
Abstract
The efficacy of osimertinib, a third-generation epidermal growth factor receptor tyrosine kinase inhibitor, has been evaluated in glioblastoma (GBM) through preclinical and clinical trials. However, the underlying mechanism of osimertinib-induced GBM cell death and the underlying resistance mechanism to osimertinib remains unclear. Here, we demonstrate that Osimertinib induces paraptosis in GBM cells, as evidenced by the formation of cytoplasmic vacuoles, accumulation of ubiquitinated proteins, and upregulation of endoplasmic reticulum (ER) stress markers like CHOP. Additionally, neither apoptosis nor autophagy was involved in the osimertinib-induced cell death. RNAseq analysis revealed ER stress was the most significantly downregulated pathway upon exposure to osimertinib. Consistently, pharmacologically targeting the PERK-eIF2α axis impaired osimertinib-induced paraptosis. Notably, we show that the expression of thyroid receptor-interacting protein 13 (TRIP13), an AAA+ATPase, alleviated osimertinib-triggered paraptosis, thus conferring resistance. Intriguingly, MK-2206, an AKT inhibitor, downregulated TRIP13 levels and synergized with Osimertinib to suppress TRIP13-induced high GBM cell growth in vitro and in vivo. Together, our findings reveal a novel mechanism of action associated with the anti-GBM effects of osimertinib involving ER stress-regulated paraptosis. Furthermore, we identify a TRIP13-driven resistance mechanism against Osimertinib in GBM and offer a combination strategy using MK-2206 to overcome such resistance.
Collapse
Affiliation(s)
- Lulu Hu
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China
- Department of Laboratory Medicine, Affiliated Qingdao Central Hospital, Qingdao University, 266000, Qingdao, China
| | - Ji Shi
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Dachuan Shen
- Department of Oncology, Affliated Zhongshan Hospital of Dalian University, 116001, Dalian, China
| | - Xingyue Zhai
- Clinical Nutrition Department, The Second Hospital of Dalian Medical University, 116023, Dalian, China
| | - Dapeng Liang
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China
| | - Jing Wang
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China
| | - Chunrui Xie
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China
| | - Zhiyu Xia
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China
| | - Jing Cui
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Feng Liu
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Sha Du
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China.
| | - Songshu Meng
- Institute of Cancer Stem Cell, Dalian Medical University, 116044, Dalian, China.
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China.
| |
Collapse
|
21
|
Mitsueda R, Toda H, Shinden Y, Fukuda K, Yasudome R, Kato M, Kikkawa N, Ohtsuka T, Nakajo A, Seki N. Oncogenic Targets Regulated by Tumor-Suppressive miR-30c-1-3p and miR-30c-2-3p: TRIP13 Facilitates Cancer Cell Aggressiveness in Breast Cancer. Cancers (Basel) 2023; 15:4189. [PMID: 37627217 PMCID: PMC10453418 DOI: 10.3390/cancers15164189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/26/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Accumulating evidence suggests that the miR-30 family act as critical players (tumor-suppressor or oncogenic) in a wide range of human cancers. Analysis of microRNA (miRNA) expression signatures and The Cancer Genome Atlas (TCGA) database revealed that that two passenger strand miRNAs, miR-30c-1-3p and miR-30c-2-3p, were downregulated in cancer tissues, and their low expression was closely associated with worse prognosis in patients with BrCa. Functional assays showed that miR-30c-1-3p and miR-30c-2-3p overexpression significantly inhibited cancer cell aggressiveness, suggesting these two miRNAs acted as tumor-suppressors in BrCa cells. Notably, involvement of passenger strands of miRNAs is a new concept of cancer research. Further analyses showed that seven genes (TRIP13, CCNB1, RAD51, PSPH, CENPN, KPNA2, and MXRA5) were putative targets of miR-30c-1-3p and miR-30c-2-3p in BrCa cells. Expression of seven genes were upregulated in BrCa tissues and predicted a worse prognosis of the patients. Among these genes, we focused on TRIP13 and investigated the functional significance of this gene in BrCa cells. Luciferase reporter assays showed that TRIP13 was directly regulated by these two miRNAs. TRIP13 knockdown using siRNA attenuated BrCa cell aggressiveness. Inactivation of TRIP13 using a specific inhibitor prevented the malignant transformation of BrCa cells. Exploring the molecular networks controlled by miRNAs, including passenger strands, will facilitate the identification of diagnostic markers and therapeutic target molecules in BrCa.
Collapse
Affiliation(s)
- Reiko Mitsueda
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (R.M.); (H.T.); (Y.S.); (K.F.); (R.Y.); (T.O.); (A.N.)
| | - Hiroko Toda
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (R.M.); (H.T.); (Y.S.); (K.F.); (R.Y.); (T.O.); (A.N.)
| | - Yoshiaki Shinden
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (R.M.); (H.T.); (Y.S.); (K.F.); (R.Y.); (T.O.); (A.N.)
| | - Kosuke Fukuda
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (R.M.); (H.T.); (Y.S.); (K.F.); (R.Y.); (T.O.); (A.N.)
| | - Ryutaro Yasudome
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (R.M.); (H.T.); (Y.S.); (K.F.); (R.Y.); (T.O.); (A.N.)
| | - Mayuko Kato
- Department of Functional Genomics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.K.); (N.K.)
| | - Naoko Kikkawa
- Department of Functional Genomics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.K.); (N.K.)
| | - Takao Ohtsuka
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (R.M.); (H.T.); (Y.S.); (K.F.); (R.Y.); (T.O.); (A.N.)
| | - Akihiro Nakajo
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan; (R.M.); (H.T.); (Y.S.); (K.F.); (R.Y.); (T.O.); (A.N.)
| | - Naohiko Seki
- Department of Functional Genomics, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (M.K.); (N.K.)
| |
Collapse
|
22
|
Kannampuzha S, Gopalakrishnan AV. Cancer chemoresistance and its mechanisms: Associated molecular factors and its regulatory role. Med Oncol 2023; 40:264. [PMID: 37550533 DOI: 10.1007/s12032-023-02138-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023]
Abstract
Cancer therapy has advanced from tradition chemotherapy methods to targeted therapy, novel drug delivery mechanisms, combination therapies etc. Although several novel chemotherapy strategies have been introduced, chemoresistance still remains as one of the major barriers in cancer treatments. Chemoresistance can lead to relapse and hinder the development of improved clinical results for cancer patients, and this continues to be the major hurdle in cancer therapy. Anticancer drugs acquire chemoresistance through different mechanisms. Understanding these mechanisms is crucial to overcome and increase the efficiency of the cancer therapies that are employed. The potential molecular pathways behind chemoresistance include tumor heterogeneity, elevated drug efflux, multidrug resistance, interconnected signaling pathways, and other factors. To surpass this limitation, new clinical tactics are to be introduced. This review aims to compile the most recent information on the molecular pathways that regulate chemoresistance in cancers, which will aid in development of new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
23
|
Ni F, Liu X, Xia Y, Zhu H, Li F, Zhang N, Xu H. TRIP 13-dependent pathways promote the development of gastric cancer. Funct Integr Genomics 2023; 23:232. [PMID: 37432513 PMCID: PMC10335954 DOI: 10.1007/s10142-023-01160-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023]
Abstract
TRIP13 is highly expressed in various human tumors and promotes tumorigenesis. We aimed to explore the biological effect of TRIP13 on gastric cancer. The RNA sequence data were retrieved from TCGA to evaluate TRIP13 mRNA expression in gastric cancer. Paired formalin-fixed paraffin-embedded blocks were further analyzed to verify the relationship between TRIP13 expression and carcinogenic status. The functions of TRIP13 on the proliferation of gastric malignancy were investigated by MTT, flow cytometry, colony formation experiment, and nude mouse tumor formation experiment. Finally, microarray analysis of TRIP13-related pathways was performed to identify the potential underlying mechanism of TRIP13 in gastric cancer. TRIP13 was found to have high expression in tumor samples. TRIP13 expression status was significantly subjective to tumor-node-metastasis (TNM) staging and poor survival. The downregulation of TRIP13 promoted apoptosis and inhibited tumor growth. TRIP13-dependent JAK/STAT and NF-κB signaling cascade were found as two key pathways in the carcinogenesis of GC. In conclusion, TRIP13 participates in the carcinogenesis of stomach cancer, and its overexpression in the cancerous tissues dovetail with advanced stage and survival. Moreover, TRIP13 functions as an upstream regulator of the JAK/STAT and p53 signaling pathways, which play critical roles in developing various malignancies.
Collapse
Affiliation(s)
- Fengming Ni
- Department of Gastroenterology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Xinmin Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yan Xia
- Department of Gastroenterology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - He Zhu
- Department of Gastroenterology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Fudong Li
- Department of Gastroenterology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Nan Zhang
- Department of Gastroenterology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China
| | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, China.
| |
Collapse
|
24
|
Ma Y, Mu X, Gao R, Zhang Y, Geng Y, Chen X, Yin X, Li F, He J. Maternal exposure to dibutyl phthalate regulates MSH6 crotonylation to impair homologous recombination in fetal oocytes. JOURNAL OF HAZARDOUS MATERIALS 2023; 455:131540. [PMID: 37167869 DOI: 10.1016/j.jhazmat.2023.131540] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/12/2023] [Accepted: 04/27/2023] [Indexed: 05/13/2023]
Abstract
Homologous recombination (HR) during early oogenesis repairs programmed double-strand breaks (DSBs) to ensure female fertility and offspring health. The exposure of fetal ovaries to endocrine disrupting chemicals (EDCs) can cause reproductive disorders in the adulthood. The EDC dibutyl phthalate (DBP) is widely distributed in flexible plastic products, leading to ubiquitous human exposure. Here, we report that maternal exposure to DBP caused gross aberrations in meiotic prophase I of fetal oocytes, including delayed progression, impaired DNA damage response, uncoupled localization of DMC1 and RAD51, and decreased HR. However, programmed DSBs were efficiently repaired. DBP exposure negatively regulated lysine crotonylation (Kcr) of MSH6. Similar meiotic defects were observed in fetal ovaries with targeted disruption of Msh6, and mutation of K544cr of MSH6 impaired its association with Ku70, thereby promoting non-homologous end joining (NHEJ) and inhibiting HR. Unlike mature F1 females, F2 female mice exhibited premature follicular activation, precocious puberty, and anxiety-like behaviors. Therefore, DBP can influence early meiotic events, and Kcr of MSH6 may regulate preferential induction of HR or NHEJ for DNA repair during meiosis.
Collapse
Affiliation(s)
- Yidan Ma
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Xinyi Mu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Rufei Gao
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Yan Zhang
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China; College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Xuemei Chen
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Yin
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Fangfang Li
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China
| | - Junlin He
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing 400016, PR China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
25
|
Gaudreau-Lapierre A, Klonisch T, Nicolas H, Thanasupawat T, Trinkle-Mulcahy L, Hombach-Klonisch S. Nuclear High Mobility Group A2 (HMGA2) Interactome Revealed by Biotin Proximity Labeling. Int J Mol Sci 2023; 24:ijms24044246. [PMID: 36835656 PMCID: PMC9966875 DOI: 10.3390/ijms24044246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/23/2023] Open
Abstract
The non-histone chromatin binding protein High Mobility Group AT-hook protein 2 (HMGA2) has important functions in chromatin remodeling, and genome maintenance and protection. Expression of HMGA2 is highest in embryonic stem cells, declines during cell differentiation and cell aging, but it is re-expressed in some cancers, where high HMGA2 expression frequently coincides with a poor prognosis. The nuclear functions of HMGA2 cannot be explained by binding to chromatin alone but involve complex interactions with other proteins that are incompletely understood. The present study used biotin proximity labeling, followed by proteomic analysis, to identify the nuclear interaction partners of HMGA2. We tested two different biotin ligase HMGA2 constructs (BioID2 and miniTurbo) with similar results, and identified known and new HMGA2 interaction partners, with functionalities mainly in chromatin biology. These HMGA2 biotin ligase fusion constructs offer exciting new possibilities for interactome discovery research, enabling the monitoring of nuclear HMGA2 interactomes during drug treatments.
Collapse
Affiliation(s)
- Antoine Gaudreau-Lapierre
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Pathology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Medical Microbiology & Infectious Diseases, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3T 2N2, Canada
- Research Institute in Oncology and Hematology (RIOH), CancerCare Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Hannah Nicolas
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Thatchawan Thanasupawat
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Laura Trinkle-Mulcahy
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3T 2N2, Canada
- Department of Pathology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: ; Tel.: +1-204-789-3982; Fax: +1-204-789-3920
| |
Collapse
|
26
|
TRIP13 Participates in Immediate-Early Sensing of DNA Strand Breaks and ATM Signaling Amplification through MRE11. Cells 2022; 11:cells11244095. [PMID: 36552858 PMCID: PMC9776959 DOI: 10.3390/cells11244095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Thyroid hormone receptor-interacting protein 13 (TRIP13) participates in various regulatory steps related to the cell cycle, such as the mitotic spindle assembly checkpoint and meiotic recombination, possibly by interacting with members of the HORMA domain protein family. Recently, it was reported that TRIP13 could regulate the choice of the DNA repair pathway, i.e., homologous recombination (HR) or nonhomologous end-joining (NHEJ). However, TRIP13 is recruited to DNA damage sites within a few seconds after damage and may therefore have another function in DNA repair other than regulation of the pathway choice. Furthermore, the depletion of TRIP13 inhibited both HR and NHEJ, suggesting that TRIP13 plays other roles besides regulation of choice between HR and NHEJ. To explore the unidentified functions of TRIP13 in the DNA damage response, we investigated its genome-wide interaction partners in the context of DNA damage using quantitative proteomics with proximity labeling. We identified MRE11 as a novel interacting partner of TRIP13. TRIP13 controlled the recruitment of MDC1 to DNA damage sites by regulating the interaction between MDC1 and the MRN complex. Consistently, TRIP13 was involved in ATM signaling amplification. Our study provides new insight into the function of TRIP13 in immediate-early DNA damage sensing and ATM signaling activation.
Collapse
|
27
|
Ghosh S, Mazumdar T, Xu W, Powell RT, Stephan C, Shen L, Shah PA, Pickering CR, Myers JN, Wang J, Frederick MJ, Johnson FM. Combined TRIP13 and Aurora Kinase Inhibition Induces Apoptosis in Human Papillomavirus-Driven Cancers. Clin Cancer Res 2022; 28:4479-4493. [PMID: 35972731 PMCID: PMC9588713 DOI: 10.1158/1078-0432.ccr-22-1627] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/11/2022] [Accepted: 08/11/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE Human papillomavirus (HPV) causes >5% of cancers, but no therapies uniquely target HPV-driven cancers. EXPERIMENTAL DESIGN We tested the cytotoxic effect of 864 drugs in 16 HPV-positive and 17 HPV-negative human squamous cancer cell lines. We confirmed apoptosis in vitro and in vivo using patient-derived xenografts. Mitotic pathway components were manipulated with drugs, knockdown, and overexpression. RESULTS Aurora kinase inhibitors were more effective in vitro and in vivo in HPV-positive than in HPV-negative models. We hypothesized that the mechanism of sensitivity involves retinoblastoma (Rb) expression because the viral oncoprotein E7 leads to Rb protein degradation, and basal Rb protein expression correlates with Aurora inhibition-induced apoptosis. Manipulating Rb directly, or by inducing E7 expression, altered cells' sensitivity to Aurora kinase inhibitors. Rb affects expression of the mitotic checkpoint genes MAD2L1 and BUB1B, which we found to be highly expressed in HPV-positive patient tumors. Knockdown of MAD2L1 or BUB1B reduced Aurora kinase inhibition-induced apoptosis, whereas depletion of the MAD2L1 regulator TRIP13 enhanced it. TRIP13 is a potentially druggable AAA-ATPase. Combining Aurora kinase inhibition with TRIP13 depletion led to extensive apoptosis in HPV-positive cancer cells but not in HPV-negative cancer cells. CONCLUSIONS Our data support a model in which HPV-positive cancer cells maintain a balance of MAD2L1 and TRIP13 to allow mitotic exit and survival in the absence of Rb. Because it does not affect cells with intact Rb function, this novel combination may have a wide therapeutic window, enabling the effective treatment of Rb-deficient cancers.
Collapse
Affiliation(s)
- Soma Ghosh
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tuhina Mazumdar
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Xu
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Reid T. Powell
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M College of Medicine, Houston, Texas
| | - Clifford Stephan
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M College of Medicine, Houston, Texas
| | - Li Shen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pooja A. Shah
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Curtis R. Pickering
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Jeffery N. Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
| | | | - Faye M. Johnson
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences, Houston, Texas
- Corresponding author. Faye M. Johnson, M.D., PhD., Faculty, Graduate School of Biomedical Sciences; Professor, Thoracic, Head and Neck Medical Oncology, University of Texas, M.D. Anderson Cancer Center, 1515 Holcombe, Box 432, Houston, TX 77030, phone 713-792-6363, fax 713-792-1220,
| |
Collapse
|
28
|
Rios Garcia M, Meissburger B, Chan J, de Guia RM, Mattijssen F, Roessler S, Birkenfeld AL, Raschzok N, Riols F, Tokarz J, Giroud M, Gil Lozano M, Hartleben G, Nawroth P, Haid M, López M, Herzig S, Berriel Diaz M. Trip13 Depletion in Liver Cancer Induces a Lipogenic Response Contributing to Plin2-Dependent Mitotic Cell Death. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104291. [PMID: 36031387 PMCID: PMC9561781 DOI: 10.1002/advs.202104291] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 08/04/2022] [Indexed: 06/15/2023]
Abstract
Aberrant energy metabolism and cell cycle regulation both critically contribute to malignant cell growth and both processes represent targets for anticancer therapy. It is shown here that depletion of the AAA+-ATPase thyroid hormone receptor interacting protein 13 (Trip13) results in mitotic cell death through a combined mechanism linking lipid metabolism to aberrant mitosis. Diminished Trip13 levels in hepatocellular carcinoma cells result in insulin-receptor-/Akt-pathway-dependent accumulation of lipid droplets, which act as functional acentriolar microtubule organizing centers disturbing mitotic spindle polarity. Specifically, the lipid-droplet-coating protein perilipin 2 (Plin2) is required for multipolar spindle formation, induction of DNA damage, and mitotic cell death. Plin2 expression in different tumor cells confers susceptibility to cell death induced by Trip13 depletion as well as treatment with paclitaxel, a spindle-interfering drug commonly used against different cancers. Thus, assessment of Plin2 levels enables the stratification of tumor responsiveness to mitosis-targeting drugs, including clinically approved paclitaxel and Trip13 inhibitors currently under development.
Collapse
|
29
|
Cao Y, Huang F, Liu J, Qi H, Xiao J. MiR-129-5p/TRIP13 affects malignant phenotypes of colorectal cancer cells. Histol Histopathol 2022; 37:879-888. [PMID: 35362548 DOI: 10.14670/hh-18-455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Aberrant miR-129-5p expression is a key modulator of cancer development. But how the miRNA affects colorectal cancer (CRC) remains unclear. This study was designed to illustrate the underlying mechanism of miR-129-5p in CRC. METHODS MiR-129-5p expression at cellular level was assayed by qRT-PCR. Its role in CRC cell phenotypes was studied by cell function experiments. The binding relationship between miR-129-5p and TRIP13 was analyzed and verified by target changed to bioinformatics prediction and dual-luciferase detection. Furthermore, the functional mechanism based on miR-129-5p and TRIP13 in CRC was studied through rescue experiments. RESULTS CRC cell lines presented prominently lower miR-129-5p levels than the normal colon epithelial cell line. The forced miR-129-5p level suppressed CRC cell growth. TRIP13 was proved to be a target of miR-129-5p in CRC cells, and miR-129-5p overexpression reduced TRIP13 expression. TRIP13 knockdown resulted in cell cycle arrest. Additionally, TRIP13 overexpression restored the impacts of miR-129-5p overexpression on cell malignant phenotypes and cell cycle. CONCLUSION MiR-129-5p down-regulated TRIP13 expression, thereby restraining the malignant progression of CRC cells. The findings may offer a new target for molecular therapy of CRC.
Collapse
Affiliation(s)
- Yongqing Cao
- Department of Hematology, The first hospital of Changsha, Changsha City, Hunan Province, China.
| | - Fang Huang
- Department of Hematology, The first hospital of Changsha, Changsha City, Hunan Province, China
| | - Jiheng Liu
- Department of Hematology, The first hospital of Changsha, Changsha City, Hunan Province, China
| | - Hui Qi
- Department of Hematology, The first hospital of Changsha, Changsha City, Hunan Province, China
| | - Jinjun Xiao
- Department of Hematology, The first hospital of Changsha, Changsha City, Hunan Province, China
| |
Collapse
|
30
|
Hu H, Zhang S, Guo J, Meng F, Chen X, Gong F, Lu G, Zheng W, Lin G. Identification of Novel Variants of Thyroid Hormone Receptor Interaction Protein 13 That Cause Female Infertility Characterized by Zygotic Cleavage Failure. Front Physiol 2022; 13:899149. [PMID: 35812326 PMCID: PMC9259851 DOI: 10.3389/fphys.2022.899149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Zygotic cleavage failure (ZCF) is a severe, early type of embryonic arrest in which zygotes cannot complete the first cleavage. Although mutations in BTG4 and CHEK1 have been identified as genetic causes of ZCF, these genes only explain a small population of ZCF cases. Thus, the underlying genetic causes for other affected individuals need to be identified. Here, we identified three TRIP13 missense variants responsible for ZCF in two patients and showed that they followed a recessive inheritance pattern. All three variants resulted in obvious changes in hydrogen bonding and consistent increase in DNA damage. Additionally, transcriptomic sequencing of oocytes and arrested embryos containing these variants suggested a greater number of differentially expressed transcripts in germinal vesicle (GV) oocytes than in 1-cell embryos. Vital genes for energy metabolism and cell cycle procession were widely and markedly downregulated, while DNA repair-related genes were significantly upregulated in both GV oocytes and 1-cell embryos of patients. These findings highlight a critical role of TRIP13 in meiosis and mitosis, as well as expand the genetic and phenotypic spectra of TR1P13 variants with respect to female infertility, especially in relation to ZCF.
Collapse
Affiliation(s)
- Huiling Hu
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
| | - Shuoping Zhang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Jing Guo
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Fei Meng
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Xueqin Chen
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
| | - Fei Gong
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Guangxiu Lu
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Wei Zheng
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- *Correspondence: Wei Zheng, ; Ge Lin,
| | - Ge Lin
- Laboratory of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, Central South University, Changsha, China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- *Correspondence: Wei Zheng, ; Ge Lin,
| |
Collapse
|
31
|
Zhang LT, Ke LX, Wu XY, Tian HT, Deng HZ, Xu LY, Li EM, Long L. TRIP13 Induces Nedaplatin Resistance in Esophageal Squamous Cell Carcinoma by Enhancing Repair of DNA Damage and Inhibiting Apoptosis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7295458. [PMID: 35601150 PMCID: PMC9115607 DOI: 10.1155/2022/7295458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/14/2022] [Accepted: 03/28/2022] [Indexed: 02/05/2023]
Abstract
Thyroid hormone receptor interactor 13 (TRIP13) plays a crucial role in poor prognosis and chemotherapy resistance of cancer patients. This present study is aimed at investigating the role of high expression of TRIP13 inducing nedaplatin (NDP) resistance in esophageal squamous cell carcinoma (ESCC) cells. High expression of TRIP13 promoted the proliferation and migration of ESCC cells performed by MTS assay, colony formation assay, wound healing assay, and transwell assay. High TRIP13 expression induced NDP resistance to ESCC based on the cell proliferation promoting/inhibition rate and cell migration promoting/inhibition rate analysis, flow cytometry assay of apoptotic subpopulations with a combination of Annexin V-FITC and propidium iodide, and Western blot analysis downregulating cleaved PARP, γH2A.X, cleaved caspase-3, and Bax and upregulating Bcl-2 expression. This study indicated that high expression of TRIP13 promoted proliferation and migration of ESCC cells and induced NDP resistance via enhancing repair of DNA damage and inhibiting apoptosis. This will provide a preliminary reference for the clinical use of NDP in ESCC treatment.
Collapse
Affiliation(s)
- Lin-Ting Zhang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| | - Li-Xin Ke
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| | - Xin-Yi Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| | - Hui-Ting Tian
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| | - Hua-Zhen Deng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| | - Li-Yan Xu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- Institute of Basic Medical Science, Cancer Research Center, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| | - Lin Long
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- Institute of Basic Medical Science, Cancer Research Center, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, 515041 Guangdong Province, China
| |
Collapse
|
32
|
Agarwal S, Afaq F, Bajpai P, Kim H, Elkholy A, Behring M, Chandrashekar DS, Diffalha SA, Khushman M, Sugandha SP, Varambally S, Manne U. DCZ0415, a small-molecule inhibitor targeting TRIP13, inhibits EMT and metastasis via inactivation of the FGFR4/STAT3 axis and the Wnt/β-catenin pathway in colorectal cancer. Mol Oncol 2022; 16:1728-1745. [PMID: 35194944 PMCID: PMC9019876 DOI: 10.1002/1878-0261.13201] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 11/11/2022] Open
Abstract
Thyroid receptor-interacting protein 13 (TRIP13), a protein of the AAA-ATPase family, is upregulated in various human cancers, including colorectal cancer (CRC). This study focused on the inhibition of TRIP13-induced CRC progression and signalling by DCZ0415, a small molecule targeting TRIP13. It demonstrated potent antitumour activity in TRIP13-deregulated cancer cell lines, regardless of their p53, KRAS, BRAF, epidermal growth factor receptor or microsatellite instability status. The treatment of CRC cells with DCZ0415 resulted in decreased cell proliferation, induced cell cycle arrest in the G2-M phase and increased apoptosis. DCZ0415 diminished xenograft tumour growth and metastasis of CRC in immunocompromised mice. DCZ0415 reduced expression of fibroblast growth factor receptor 4 (FGFR4), signal transducer and activator of transcription 3 (STAT3), and proteins associated with the epithelial-mesenchymal transition and nuclear factor kappa B (NF-κB) pathways in cells and xenografts exhibiting high expression of TRIP13. Additionally, DCZ0415 decreased cyclin D1, β-catenin and T-cell factor 1, leading to the inactivation of the Wnt/β-catenin pathway. In a syngeneic CRC model, DCZ0415 treatment induced an immune response by decreasing PD1 and CTLA4 levels and increasing granzyme B, perforin and interferon gamma. In sum, DCZ04145 inhibits the TRIP13-FGFR4-STAT3 axis, inactivates NF-κB and Wnt/β-catenin signalling, activates antitumour immune response and reduces the progression and metastasis of CRC. This study provides a rationale to evaluate DCZ0415 clinically for the treatment of a subset of CRCs that exhibit dysregulated TRIP13 and FGFR4.
Collapse
Affiliation(s)
- Sumit Agarwal
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Farrukh Afaq
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Prachi Bajpai
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Hyung‐Gyoon Kim
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Amr Elkholy
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Michael Behring
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | | | - Sameer Al Diffalha
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| | - Moh’d Khushman
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
- Department of MedicineDivision of Medical OncologyUniversity of Alabama at BirminghamALUSA
| | - Shajan P. Sugandha
- Department of MedicineDivision of GastroenterologyUniversity of Alabama at BirminghamALUSA
| | - Sooryanarayana Varambally
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| | - Upender Manne
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- O’Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| |
Collapse
|
33
|
Lan J, Huang J, Tao X, Gao Y, Zhang L, Huang W, Luo J, Liu C, Deng Y, Liu L, Liu X. Evaluation of the TRIP13 level in breast cancer and insights into potential molecular pathways. J Cell Mol Med 2022; 26:2673-2685. [PMID: 35322916 PMCID: PMC9077308 DOI: 10.1111/jcmm.17278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/10/2022] [Accepted: 02/27/2022] [Indexed: 11/29/2022] Open
Abstract
TRIP13 is a member of the large superfamily of the AAA + ATPase proteins and is associated with a variety of activities. Emerging evidence has shown that TRIP13 may serve as an oncogene. However, the function of TRIP13 in breast cancer (BC) has not yet been elucidated. Here, a variety of bioinformatic tools and laboratory experiments were combined to analyse the expression patterns, prognostic value and functional network of TRIP13 in BC. Multiple databases and immunohistochemistry (IHC) indicated a higher TRIP13 expression in BC tissue compared with normal tissue. TRIP13 was highly expressed in lung metastatic lesions compared with primary tumours in a 4T1 cell implantation BALB/c mouse model of BC. Kaplan–Meier plots also revealed that high TRIP13 expression correlated with poor survival in patients with BC. Furthermore, gene set enrichment analysis revealed that TRIP13 was primarily enriched in the signalling pathway of PI3K‐AKT‐mTOR. Suppressing TRIP13 could inhibit the expression of related genes, as well as the proliferation and migration of BC cell. Finally, 10 hub genes with a high score of connectivity were filtered from the protein–protein interaction (PPI) network, including MAD2L1, CDC20, CDC5L, CDK1, CCNA2, BUB1B, RAD51, SPO11, KIF11 and AURKB. Thus, TRIP13 may be a promising prognostic biomarker and an effective therapeutic target for BC.
Collapse
Affiliation(s)
- Jin Lan
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jingzhan Huang
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xinyi Tao
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yuan Gao
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Longshan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiqiang Huang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Luo
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Chuqin Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Yunyao Deng
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Lixin Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaolong Liu
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
Impact of Non-Coding RNAs on Chemotherapeutic Resistance in Oral Cancer. Biomolecules 2022; 12:biom12020284. [PMID: 35204785 PMCID: PMC8961659 DOI: 10.3390/biom12020284] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Drug resistance in oral cancer is one of the major problems in oral cancer therapy because therapeutic failure directly results in tumor recurrence and eventually in metastasis. Accumulating evidence has demonstrated the involvement of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in processes related to the development of drug resistance. A number of studies have shown that ncRNAs modulate gene expression at the transcriptional or translational level and regulate biological processes, such as epithelial-to-mesenchymal transition, apoptosis, DNA repair and drug efflux, which are tightly associated with drug resistance acquisition in many types of cancer. Interestingly, these ncRNAs are commonly detected in extracellular vesicles (EVs) and are known to be delivered into surrounding cells. This intercellular communication via EVs is currently considered to be important for acquired drug resistance. Here, we review the recent advances in the study of drug resistance in oral cancer by mainly focusing on the function of ncRNAs, since an increasing number of studies have suggested that ncRNAs could be therapeutic targets as well as biomarkers for cancer diagnosis.
Collapse
|
35
|
TRIP13, identified as a hub gene of tumor progression, is the target of microRNA-4693-5p and a potential therapeutic target for colorectal cancer. Cell Death Dis 2022; 8:35. [PMID: 35075117 PMCID: PMC8786872 DOI: 10.1038/s41420-022-00824-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 12/18/2021] [Accepted: 01/06/2022] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the digestive tract malignancies whose early symptoms are not obvious. This study aimed to identify novel targets for CRC therapy, especially early-stage CRC, by reanalyzing the publicly available GEO and TCGA databases. Thyroid hormone receptor interactor 13 (TRIP13) correlated with tumor progression and prognosis of patients after several rounds of analysis, including weighted gene correlation network analysis (WGCNA), and further chosen for experimental validation in cancer cell lines and patient samples. We identified that mRNA and protein levels of TRIP13 increased in CRC cells and tumor tissues with tumor progression. miR-4693-5p was significantly downregulated in CRC tumor tissues and bound to the 3′ untranslated region (3′UTR) of TRIP13, downregulating TRIP13 expression. DCZ0415, a small molecule inhibitor targeting TRIP13, induced anti-tumor activity in vitro and in vivo. DCZ0415 markedly suppressed CRC cell proliferation, migration, and tumor growth, promoted cell apoptosis, and resulted in the arrest of the cell cycle. Our research suggests that TRIP13 might play a crucial role in CRC progression and could be a potential target for CRC therapy.
Collapse
|
36
|
Gu Y, Desai A, Corbett KD. Evolutionary Dynamics and Molecular Mechanisms of HORMA Domain Protein Signaling. Annu Rev Biochem 2022; 91:541-569. [PMID: 35041460 DOI: 10.1146/annurev-biochem-090920-103246] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Controlled assembly and disassembly of multi-protein complexes is central to cellular signaling. Proteins of the widespread and functionally diverse HORMA family nucleate assembly of signaling complexes by binding short peptide motifs through a distinctive safety-belt mechanism. HORMA proteins are now understood as key signaling proteins across kingdoms, serving as infection sensors in a bacterial immune system and playing central roles in eukaryotic cell cycle, genome stability, sexual reproduction, and cellular homeostasis pathways. Here, we describe how HORMA proteins' unique ability to adopt multiple conformational states underlies their functions in these diverse contexts. We also outline how a dedicated AAA+ ATPase regulator, Pch2/TRIP13, manipulates HORMA proteins' conformational states to activate or inactivate signaling in different cellular contexts. The emergence of Pch2/TRIP13 as a lynchpin for HORMA protein action in multiple genome-maintenance pathways accounts for its frequent misregulation in human cancers and highlights TRIP13 as a novel therapeutic target. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Yajie Gu
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, California, USA;
| | - Arshad Desai
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, California, USA; .,Section of Cell & Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, USA.,Ludwig Institute for Cancer Research, San Diego Branch, La Jolla, California, USA
| | - Kevin D Corbett
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
37
|
Phosphorylation of TRIP13 at Y56 induces radiation resistance but sensitizes head and neck cancer to cetuximab. Mol Ther 2022; 30:468-484. [PMID: 34111559 PMCID: PMC8753291 DOI: 10.1016/j.ymthe.2021.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/10/2021] [Accepted: 06/01/2021] [Indexed: 01/07/2023] Open
Abstract
Radiation therapy, a mainstay of treatment for head and neck cancer, is not always curative due to the development of treatment resistance; additionally, multi-institutional trials have questioned the efficacy of concurrent radiation with cetuximab, the epidermal growth factor receptor (EGFR) inhibitor. We unraveled a mechanism for radiation resistance; that is, radiation induces EGFR, which phosphorylates TRIP13 (thyroid hormone receptor interactor 13) on tyrosine 56. Phosphorylated (phospho-)TRIP13 promotes non-homologous end joining (NHEJ) repair to induce radiation resistance. NHEJ is the main repair pathway for radiation-induced DNA damage. Tumors expressing high TRIP13 do not respond to radiation but are sensitive to cetuximab or cetuximab combined with radiation. Suppression of phosphorylation of TRIP13 at Y56 abrogates these effects. These findings show that EGFR-mediated phosphorylation of TRIP13 at Y56 is a vital mechanism of radiation resistance. Notably, TRIP13-pY56 could be used to predict the response to radiation or cetuximab and could be explored as an actionable target.
Collapse
|
38
|
Hama T, Nagesh PK, Chowdhury P, Moore BM, Yallapu MM, Regner KR, Park F. DNA damage is overcome by TRIP13 overexpression during cisplatin nephrotoxicity. JCI Insight 2021; 6:139092. [PMID: 34806647 PMCID: PMC8663775 DOI: 10.1172/jci.insight.139092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent to treat a wide array of cancers that is frequently associated with toxic injury to the kidney due to oxidative DNA damage and perturbations in cell cycle progression leading to cell death. In this study, we investigated whether thyroid receptor interacting protein 13 (TRIP13) plays a central role in the protection of the tubular epithelia following cisplatin treatment by circumventing DNA damage. Following cisplatin treatment, double-stranded DNA repair pathways were inhibited using selective blockers to proteins involved in either homologous recombination or non-homologous end joining. This led to increased blood markers of acute kidney injury (AKI) (creatinine and neutrophil gelatinase–associated lipocalin), tubular damage, activation of DNA damage marker (γ-H2AX), elevated appearance of G2/M blockade (phosphorylated histone H3 Ser10 and cyclin B1), and apoptosis (cleaved caspase-3). Conditional proximal tubule–expressing Trip13 mice were observed to be virtually protected from the cisplatin nephrotoxicity by restoring most of the pathological phenotypes back toward normal conditions. Our findings suggest that TRIP13 could circumvent DNA damage in the proximal tubules during cisplatin injury and that TRIP13 may constitute a new therapeutic target in protecting the kidney from nephrotoxicants and reduce outcomes leading to AKI.
Collapse
Affiliation(s)
- Taketsugu Hama
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Prashanth Kb Nagesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Bob M Moore
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Kevin R Regner
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
39
|
MAD2L2 dimerization and TRIP13 control shieldin activity in DNA repair. Nat Commun 2021; 12:5421. [PMID: 34521823 PMCID: PMC8440562 DOI: 10.1038/s41467-021-25724-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/28/2021] [Indexed: 12/13/2022] Open
Abstract
MAD2L2 (REV7) plays an important role in DNA double-strand break repair. As a member of the shieldin complex, consisting of MAD2L2, SHLD1, SHLD2 and SHLD3, it controls DNA repair pathway choice by counteracting DNA end-resection. Here we investigated the requirements for shieldin complex assembly and activity. Besides a dimerization-surface, HORMA-domain protein MAD2L2 has the extraordinary ability to wrap its C-terminus around SHLD3, likely creating a very stable complex. We show that appropriate function of MAD2L2 within shieldin requires its dimerization, mediated by SHLD2 and accelerating MAD2L2-SHLD3 interaction. Dimerization-defective MAD2L2 impairs shieldin assembly and fails to promote NHEJ. Moreover, MAD2L2 dimerization, along with the presence of SHLD3, allows shieldin to interact with the TRIP13 ATPase, known to drive topological switches in HORMA-domain proteins. We find that appropriate levels of TRIP13 are important for proper shieldin (dis)assembly and activity in DNA repair. Together our data provide important insights in the dependencies for shieldin activity.
Collapse
|
40
|
Liu X, Shen X, Zhang J. TRIP13 exerts a cancer-promoting role in cervical cancer by enhancing Wnt/β-catenin signaling via ACTN4. ENVIRONMENTAL TOXICOLOGY 2021; 36:1829-1840. [PMID: 34061428 DOI: 10.1002/tox.23303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/10/2021] [Accepted: 05/24/2021] [Indexed: 06/12/2023]
Abstract
Increasing evidence has indicated that thyroid hormone receptor interacting protein 13 (TRIP13) exerts a cancer-promoting role in a broad spectrum of cancers. However, the detailed relevance and function of TRIP13 in cervical cancer remain undefined. The goal of this work was to evaluate the functional significance and mechanism of TRIP13 in cervical cancer. Our data demonstrated that TRIP13 expression was markedly increased in cervical cancer tissue, and high expression of TRIP13 predicted a low survival rate in cervical cancer patients. Knockdown of TRIP13 caused a significant reduction in the proliferation and invasion of cervical cancer cells. By contrast, over-expression of TRIP13 accelerated the proliferation and invasion of cervical cancer cells. Further data revealed that TRIP13 enhanced the activation of Wnt/β-catenin signaling associated with modulation of α-Actinin-4 (ACTN4). Knockdown of ACTN4 markedly reversed TRIP13-mediated activation of Wnt/β-catenin signaling. In addition, inhibition of Wnt/β-catenin signaling reversed TRIP13-induced cancer-promoting effects in cervical cancer cells. Knockdown of TRIP13 markedly retarded the tumor formation and growth of cervical cells in vivo in nude mice. Taken together, the data of this work indicate that TRIP13 accelerates the proliferation and invasion of cervical cancer by enhancing Wnt/β-catenin signaling via regulation of ACTN4. These findings underscore a relevance of the TRIP13/ACTN4/Wnt/β-catenin signaling axis in the progression of cervical cancer and suggest TRIP13 as a potential target for treatment of cervical cancer.
Collapse
Affiliation(s)
- Xiaoying Liu
- Department of Gynecology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, China
| | - Xin Shen
- Department of Gynecology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, China
| | - Jing Zhang
- Department of Gynecology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, China
| |
Collapse
|
41
|
Li C, Xia J, Franqui-Machin R, Chen F, He Y, Ashby TC, Teng F, Xu H, Liu D, Gai D, Johnson SK, van Rhee F, Janz S, Shaughnessy JD, Tricot G, Frech I, Zhan F. TRIP13 modulates protein deubiquitination and accelerates tumor development and progression of B cell malignancies. J Clin Invest 2021; 131:e146893. [PMID: 34061780 DOI: 10.1172/jci146893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/25/2021] [Indexed: 12/22/2022] Open
Abstract
Multiple myeloma (MM), a terminally differentiated B cell malignancy, remains difficult to cure. Understanding the molecular mechanisms underlying the progression of MM may identify therapeutic targets and lead to a fundamental shift in treatment of the disease. Deubiquitination, like ubiquitination, is a highly regulated process, implicated in almost every cellular process. Multiple deubiquitinating enzymes (DUBs) have been identified, but their regulation is poorly defined. Here, we determined that TRIP13 increases cellular deubiquitination. Overexpression of TRIP13 in mice and cultured cells resulted in excess cellular deubiquitination by enhancing the association of the DUB USP7 with its substrates. We show that TRIP13 is an oncogenic protein because it accelerates B cell tumor development in transgenic mice. TRIP13-induced resistance to proteasome inhibition can be overcome by a USP7 inhibitor in vitro and in vivo. These findings suggest that TRIP13 expression plays a critical role in B cell lymphoma and MM by regulating deubiquitination of critical oncogenic (NEK2) and tumor suppressor (PTEN, p53) proteins. High TRIP13 identifies a high-risk patient group amenable to adjuvant anti-USP7 therapy.
Collapse
Affiliation(s)
- Can Li
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiliang Xia
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | | | - Fangping Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanjuan He
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Timothy Cody Ashby
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Feixiang Teng
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Hongwei Xu
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Dingxiao Liu
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Dongzheng Gai
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sarah K Johnson
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Frits van Rhee
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Siegfried Janz
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - John D Shaughnessy
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Guido Tricot
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ivana Frech
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
42
|
Zeng L, Liu YM, Yang N, Zhang T, Xie H. Hsa_circRNA_100146 Promotes Prostate Cancer Progression by Upregulating TRIP13 via Sponging miR-615-5p. Front Mol Biosci 2021; 8:693477. [PMID: 34307457 PMCID: PMC8292639 DOI: 10.3389/fmolb.2021.693477] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/17/2021] [Indexed: 01/13/2023] Open
Abstract
Objective: This study was conducted for investigating the functions of circular RNA circRNA_100146 (circRNA_100146) in the development of prostate cancer (PCa) and identifying the underlying mechanisms of the circRNA_100146/miR-615-5p/TRIP13 axis. Materials and Methods: Under the support of RT-PCR, the expression of circRNA_100146 in PCa cells was examined. Cell Counting Kit-8 (CCK-8) assays and clone formation assays were applied to the assessment of cell proliferation. We then determined cell invasion and migration through transwell assays and wound healing assays. RNA pull-down assays and luciferase reporter assays were performed for the exploration of the regulatory effects of potential molecules on the expressions of the targeting genes. In addition, a nude mouse xenograft model was applied to demonstrate the oncogenic roles of circRNA_100146 in PCa. Results: CircRNA_100146 expression was distinctly upregulated in PCa cells. Silencing of circRNA_100146 suppressed PCa cells' invasion, migration, and proliferation. CircRNA_100146 sponged miR-615-5p to suppress its expressions, while miR-615-5p targeted the 3'-UTR of TRIP13 to repress the expression of TRIP13. In addition, we observed that knockdown of miR-615-5p reversed the suppression of circRNA_100146 silence on the proliferation and invasion of PCa cells. In addition, the tumor growth was also suppressed by silencing circRNA_100146 in vivo. Conclusion: CircRNA_100146 is a tumor promoter in PCa, which promoted progression by mediating the miR-615-5p/TRIP13. CircRNA_100146 can be a potential candidate for targeted therapy of PCa.
Collapse
Affiliation(s)
- Liang Zeng
- Emergency Department, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Yi-Min Liu
- Department of Anesthesiology, The Affiliated Nanhua Hospital, University of South China, Engyang, China
| | - Ning Yang
- Department of Urology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Tao Zhang
- Department of Urology, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Huang Xie
- Department of Urology, The Second Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
43
|
Singh P, Banerjee R, Piao S, Costa de Medeiros M, Bellile E, Liu M, Damodaran Puthiya Veettil D, Schmitd LB, Russo N, Danella E, Inglehart RC, Pineault KM, Wellik DM, Wolf G, D’Silva NJ. Squamous cell carcinoma subverts adjacent histologically normal epithelium to promote lateral invasion. J Exp Med 2021; 218:e20200944. [PMID: 33835136 PMCID: PMC8042603 DOI: 10.1084/jem.20200944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 01/04/2021] [Accepted: 02/16/2021] [Indexed: 12/26/2022] Open
Abstract
Recurrent and new tumors, attributed in part to lateral invasion, are frequent in squamous cell carcinomas and lead to poor survival. We identified a mechanism by which cancer subverts adjacent histologically normal epithelium to enable small clusters of cancer cells to burrow undetected under adjacent histologically normal epithelium. We show that suppression of DMBT1 within cancer promotes aggressive invasion and metastasis in vivo and is associated with metastasis in patients. Cancer cells via TGFβ1 and TNFα also suppress DMBT1 in adjacent histologically normal epithelium, thereby subverting it to promote invasion of a small population of tumor cells. The sufficiency of DMBT1 in this process is demonstrated by significantly higher satellite tumor nests in Dmbt1-/- compared with wild-type mice. Moreover, in patients, invasion of small tumor nests under adjacent histologically normal epithelium is associated with increased risk for recurrence and shorter disease-free survival. This study demonstrates a crucial role of adjacent histologically normal epithelium in invasion and its important role in the tumor microenvironment and opens new possibilities for therapeutic strategies that reduce tumor recurrence.
Collapse
Affiliation(s)
- Priyanka Singh
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Rajat Banerjee
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Songlin Piao
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Marcell Costa de Medeiros
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Emily Bellile
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Min Liu
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | | | - Ligia B. Schmitd
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Nickole Russo
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Erika Danella
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Ronald C. Inglehart
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Kyriel M. Pineault
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Greg Wolf
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI
| | - Nisha J. D’Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
44
|
Gao C, Jin G, Forbes E, Mangala LS, Wang Y, Rodriguez-Aguayo C, Amero P, Bayraktar E, Yan Y, Lopez-Berestein G, Broaddus RR, Sood AK, Xue F, Zhang W. Inactivating Mutations of the IK Gene Weaken Ku80/Ku70-Mediated DNA Repair and Sensitize Endometrial Cancer to Chemotherapy. Cancers (Basel) 2021; 13:2487. [PMID: 34065218 PMCID: PMC8160817 DOI: 10.3390/cancers13102487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/05/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
IK is a mitotic factor that promotes cell cycle progression. Our previous investigation of 271 endometrial cancer (EC) samples from the Cancer Genome Atlas (TCGA) dataset showed IK somatic mutations were enriched in a cluster of patients with high-grade and high-stage cancers, and this group had longer survival. This study provides insight into how IK somatic mutations contribute to EC pathophysiology. We analyzed the somatic mutational landscape of IK gene in 547 EC patients using expanded TCGA dataset. Co-immunoprecipitation and mass spectrometry were used to identify protein interactions. In vitro and in vivo experiments were used to evaluate IK's role in EC. The patients with IK-inactivating mutations had longer survival during 10-year follow-up. Frameshift and stop-gain were common mutations and were associated with decreased IK expression. IK knockdown led to enrichment of G2/M phase cells, inactivation of DNA repair signaling mediated by heterodimerization of Ku80 and Ku70, and sensitization of EC cells to cisplatin treatment. IK/Ku80 mutations were accompanied by higher mutation rates and associated with significantly better overall survival. Inactivating mutations of IK gene and loss of IK protein expression were associated with weakened Ku80/Ku70-mediated DNA repair, increased mutation burden, and better response to chemotherapy in patients with EC.
Collapse
Affiliation(s)
- Chao Gao
- Department of Cancer Biology, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA; (C.G.); (G.J.); (E.F.)
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; (Y.W.); (Y.Y.)
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin 300052, China
| | - Guangxu Jin
- Department of Cancer Biology, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA; (C.G.); (G.J.); (E.F.)
| | - Elizabeth Forbes
- Department of Cancer Biology, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA; (C.G.); (G.J.); (E.F.)
| | - Lingegowda S. Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (L.S.M.); (E.B.); (A.K.S.)
- Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (C.R.-A.); (G.L.-B.)
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; (Y.W.); (Y.Y.)
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin 300052, China
| | - Cristian Rodriguez-Aguayo
- Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (C.R.-A.); (G.L.-B.)
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Emine Bayraktar
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (L.S.M.); (E.B.); (A.K.S.)
- Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (C.R.-A.); (G.L.-B.)
| | - Ye Yan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; (Y.W.); (Y.Y.)
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin 300052, China
| | - Gabriel Lopez-Berestein
- Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (C.R.-A.); (G.L.-B.)
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Russell R. Broaddus
- Department of Pathology & Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA;
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (L.S.M.); (E.B.); (A.K.S.)
- Center for RNA Interference and Non-Coding RNAs, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (C.R.-A.); (G.L.-B.)
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, China; (Y.W.); (Y.Y.)
- Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin 300052, China
| | - Wei Zhang
- Department of Cancer Biology, Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA; (C.G.); (G.J.); (E.F.)
| |
Collapse
|
45
|
Chen SH, Lin HH, Li YF, Tsai WC, Hueng DY. Clinical Significance and Systematic Expression Analysis of the Thyroid Receptor Interacting Protein 13 (TRIP13) as Human Gliomas Biomarker. Cancers (Basel) 2021; 13:cancers13102338. [PMID: 34066132 PMCID: PMC8150328 DOI: 10.3390/cancers13102338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/08/2021] [Indexed: 12/20/2022] Open
Abstract
The prognosis of malignant gliomas such as glioblastoma multiforme (GBM) has remained poor due to limited therapeutic strategies. Thus, it is pivotal to determine prognostic factors for gliomas. Thyroid Receptor Interacting Protein 13 (TRIP13) was found to be overexpressed in several solid tumors, but its role and clinical significance in gliomas is still unclear. Here, we conducted a comprehensive expression analysis of TRIP13 to determine the prognostic values. Gene expression profiles of the Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA) and GSE16011 dataset showed increased TRIP13 expression in advanced stage and worse prognosis in IDH-wild type lower-grade glioma. We performed RT-PCR and Western blot to validate TRIP13 mRNA expression and protein levels in GBM cell lines. TRIP13 co-expressed genes via database screening were regulated by essential cancer-related upstream regulators (such as TP53 and FOXM1). Then, TCGA analysis revealed that more TRIP13 promoter hypomethylation was observed in GBM than in low-grade glioma. We also inferred that the upregulated TRIP13 levels in gliomas could be regulated by dysfunction of miR-29 in gliomas patient cohorts. Moreover, TRIP13-expressing tumors not only had higher aneuploidy but also tended to reduce the ratio of CD8+/Treg, which led to a worse survival outcome. Overall, these findings demonstrate that TRIP13 has with multiple functions in gliomas, and they may be crucial for therapeutic potential.
Collapse
Affiliation(s)
- Ssu-Han Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
| | - Hong-Han Lin
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan;
| | - Yao-Feng Li
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-F.L.); (W.-C.T.)
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; (Y.-F.L.); (W.-C.T.)
| | - Dueng-Yuan Hueng
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Department of Biochemistry, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: ; Tel.: +886-2-8792-3100 (ext. 18802)
| |
Collapse
|
46
|
Söylemez Z, Arıkan ES, Solak M, Arıkan Y, Tokyol Ç, Şeker H. Investigation of the expression levels of CPEB4, APC, TRIP13, EIF2S3, EIF4A1, IFNg, PIK3CA and CTNNB1 genes in different stage colorectal tumors. Turk J Med Sci 2021; 51:661-674. [PMID: 33237662 PMCID: PMC8208508 DOI: 10.3906/sag-2010-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/24/2020] [Indexed: 12/24/2022] Open
Abstract
Background/aim The aim of the study is to assess expression levels of CPEB4, APC, TRIP13, EIF2S3, EIF4A1, IFNg, PIK3CA and CTNNB1 genes in tumors and peripheral bloods of colorectal cancer patients in stages I–IV. Materials and methods The mRNA levels of the genes were determined in tumor tissues and peripheral blood samples of 45 colorectal cancer patients and colon tissues and peripheral blood samples of 5 healthy individuals. Real-time polymerase chain reaction method was used for the analysis. Results The mRNA level of the CPEB4 gene was significantly downregulated in colorectal tumor tissues and was upregulated in the peripheral blood of colorectal cancer patients relative to the controls (P < 0.05). APC mRNA level was significantly downregulated in tissues and upregulated in the peripheral blood (P < 0.05). TRIP13 mRNA level was upregulated in peripheral blood and also significantly upregulated in colorectal tumor tissues (P < 0.05). EIF2S3 mRNA level was upregulated in tissues and also significantly upregulated in peripheral blood (P < 0.05). PIK3CA mRNA level was downregulated in tissues and upregulated in peripheral blood. EIF4A1 mRNA level was downregulated in tissues and significantly upregulated in peripheral blood (P < 0.05). CTNNB1 mRNA level was downregulated in tissues and upregulated in peripheral blood. IFNg mRNA level was upregulated in both colorectal cancer tumor tissues and peripheral blood. Conclusion: TRIP13 and CPEB4 mRNA up regulation in the peripheral blood of patients with colorectal cancer may be a potential target for early stage diagnosis. In addition to this evaluation, although there is not much study on EIF2S3 and EIF4A1 mRNA changes in cases with colorectal cancer, upregulation in peripheral blood draws attention in our study. These data will shed light on the new comprehensive studies.
Collapse
Affiliation(s)
- Zafer Söylemez
- Department of Medical Biology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Evrim Suna Arıkan
- Department of Medical Biology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Mustafa Solak
- Department of Medical Genetic, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Yüksel Arıkan
- General Surgery Department, Park Hayat Hospital, Afyonkarahisar, Turkey
| | - Çiğdem Tokyol
- Department of Patology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey
| | - Hüseyin Şeker
- School of Computing and Digital Technologies, Staffordshire University, Stroke-on-Trent, United Kingdom
| |
Collapse
|
47
|
Zhang Q, Yuan J, Liu Y, Liu X, Lv T, Zhou K, Song Y. KIAA0101 knockdown inhibits cell proliferation and induces cell cycle arrest and cell apoptosis in chronic lymphocytic leukemia cells. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:487. [PMID: 33850884 PMCID: PMC8039647 DOI: 10.21037/atm-21-626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Chronic lymphocytic leukemia (CLL) is a heterogeneous disease with intense cytogenetic aberrations. Importantly, our recent report indicated that thyroid hormone receptor interactor 13 (TRIP13) is a potential new therapeutic target in CLL. In this study, we predicted 20 TRIP13-related genes and found that KIAA0101 is a novel gene that regulates cell proliferation and the cell cycle of CLL cells. Methods CD19+ B cells were isolated from the peripheral blood of 26 CLL patients and 6 healthy donors through magnetic cell sorting. Cell proliferation was assessed by the CCK-8 assay. The mRNA and protein levels of genes were examined through RT-qPCR and western blot assays, respectively. Cell cycle and cell apoptosis were measured through Annexin V-based flow cytometry and the caspase 3/7 activity assay. Potential targets of KIAA0101 were identified through microarray analysis. 20 TRIP13 related genes was predicted by Ingenuity Pathway Analysis (IPA). KIAA0101-regulated functions and molecular pathways were predicted through IPA. Results KIAA0101 knockdown had the strongest inhibitory effect on CLL cell proliferation among the 20 TRIP13-related genes. KIAA0101 was highly expressed in CD19+ B cells of CLL patients. KIAA0101 knockdown induced cell cycle arrest and cell apoptosis, and inhibited FOXO1, MYD88, and TLR4 expression in CLL cells. Conclusions Taken together, we demonstrated that KIAA0101 plays a critical role in cell proliferation and the cell cycle of human CLL cells. KIAA0101 knockdown induced cell apoptosis, and reduced FOXO1, MYD88, and TLR4 expression, and may therefore be used as a therapeutic target of CLL.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Jingjing Yuan
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yanyan Liu
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xingchen Liu
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Tianxin Lv
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Keshu Zhou
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
48
|
He S, Li G, Schätzlein AG, Humphrey PA, Weiss RM, Uchegbu IF, Martin DT. Down-regulation of GP130 signaling sensitizes bladder cancer to cisplatin by impairing Ku70 DNA repair signaling and promoting apoptosis. Cell Signal 2021; 81:109931. [PMID: 33529758 DOI: 10.1016/j.cellsig.2021.109931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/06/2021] [Accepted: 01/19/2021] [Indexed: 10/22/2022]
Abstract
Chemoresistance is one of the barriers for the development of bladder cancer treatments. Previously, we showed that glycoprotein-130 (GP130) is overexpressed in chemoresistant bladder cancer cells and that knocking down GP130 expression reduced cell viability. In our current work, we showed that down-regulation of GP130 sensitized bladder cancer cells to cisplatin-based chemotherapy by activating DNA repair signaling. We performed immunohistochemistry and demonstrated a positive correlation between the levels of Ku70, an initiator of canonical non-homologous end joining repair (c-NHEJ) and suppressor of apoptosis, and GP130 in human bladder cancer specimens. GP130 knockdown by SC144, a small molecule inhibitor, in combination with cisplatin, increased the number of DNA lesions, specifically DNA double-stranded breaks, with a subsequent increase in apoptosis and reduced cell viability. Furthermore, GP130 inhibition attenuated Ku70 expression in bladder and breast cancer cells as well as in transformed kidney cells. In addition, we fabricated a novel polymer-lipid hybrid delivery system to facilitate GP130 siRNA delivery that had a similar efficiency when compared with Lipofectamine, but induced less toxicity.
Collapse
Affiliation(s)
- Shanshan He
- Department of Urology, Yale University, New Haven, CT, USA
| | - Gang Li
- Department of Urology, Yale University, New Haven, CT, USA
| | | | | | - Robert M Weiss
- Department of Urology, Yale University, New Haven, CT, USA
| | | | | |
Collapse
|
49
|
Toden S, Zumwalt TJ, Goel A. Non-coding RNAs and potential therapeutic targeting in cancer. Biochim Biophys Acta Rev Cancer 2021; 1875:188491. [PMID: 33316377 PMCID: PMC7856203 DOI: 10.1016/j.bbcan.2020.188491] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022]
Abstract
Recent advances have begun to clarify the physiological and pathological roles of non-coding RNAs (ncRNAs) in various diseases, including cancer. Among these, microRNAs (miRNAs) have been the most studied and have emerged as key players that are involved in the regulation of important growth regulatory pathways in cancer pathogenesis. The ability of a single ncRNA to modulate the expression of multiple downstream gene targets and associated pathways, have provided a rationale to pursue them for therapeutic drug development in cancer. In this context, early data from pre-clinical studies have demonstrated that synthetic miRNA-based therapeutic molecules, along with various protective coating approaches, has allowed for their efficient delivery and anti-tumor activity. In fact, some of the miRNA-based cancer therapeutic strategies have shown promising results even in early-phase human clinical trials. While the enthusiasm for ncRNA-based cancer therapeutics continue to evolve, the field is still in the midst of unraveling a more precise understanding of the molecular mechanisms and specific downstream therapeutic targets of other lesser studied ncRNAs such as the long-non-coding RNAs, transfer RNAs, circular RNAs, small nucleolar RNAs, and piwi-interacting RNAs. This review article provides the current state of knowledge and the evolving principles for ncRNA-based therapeutic approaches in cancer, and specifically highlights the importance of data to date and the approaches that are being developed to overcome the challenges associated with their delivery and mitigating the off-target effects in human cancers.
Collapse
Affiliation(s)
- Shusuke Toden
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA
| | - Timothy J Zumwalt
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA
| | - Ajay Goel
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute and Charles A. Sammons Cancer Center, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA; Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
50
|
Agarwal S, Behring M, Kim H, Chandrashekar DS, Chakravarthi BVSK, Gupta N, Bajpai P, Elkholy A, Al Diffalha S, Datta PK, Heslin MJ, Varambally S, Manne U. TRIP13 promotes metastasis of colorectal cancer regardless of p53 and microsatellite instability status. Mol Oncol 2020; 14:3007-3029. [PMID: 33037736 PMCID: PMC7718953 DOI: 10.1002/1878-0261.12821] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/31/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Overexpression of TRIP13, a member of the AAA-ATPase family, is linked with various cancers, but its role in metastasis is unknown in colorectal cancer (CRC). In the current study, we investigated the role TRIP13 in experimental metastasis and its involvement in regulation of WNT/β-catenin and EGFR signaling pathways. Evaluation of formalin-fixed paraffin-embedded (FFPE) and frozen tissues of adenomas and CRCs, along with their corresponding normal samples, showed that TRIP13 was gradually increased in its phenotypic expression from adenoma to carcinoma and that its overexpression in CRCs was independent of patient's gender, age, race/ethnicity, pathologic stage, and p53 and microsatellite instability (MSI) status. Moreover, liver metastases of CRCs showed TRIP13 overexpression as compared to matched adjacent liver tissues, indicating the biological relevance of TRIP13 in CRC progression and metastasis. TRIP13 knockdown impeded colony formation, invasion, motility, and spheroid-forming capacity of CRC cells irrespective of their p53 and MSI status. Furthermore, xenograft studies demonstrated high expression of TRIP13 contributed to tumor growth and metastasis. Depletion of TRIP13 in CRC cells decreased metastasis and it was independent of the p53 and MSI status. Furthermore, TRIP13 interacted with a tyrosine kinase, FGFR4; this interaction could be essential for activation of the EGFR-AKT pathway. In addition, we demonstrated the involvement of TRIP13 in the Wnt signaling pathway and in the epithelial-mesenchymal transition. Cell-based assays revealed that miR-192 and PNPT1 regulate TRIP13 expression in CRC. Additionally, RNA sequencing of CRC cells with TRIP13 knockdown identified COL6A3, TREM2, SHC3, and KLK7 as downstream targets that may have functional relevance in TRIP13-mediated tumor growth and metastasis. In summary, our results demonstrated that TRIP13 promotes tumor growth and metastasis regardless of p53 and MSI status, and indicated that it is a target for therapy of CRC.
Collapse
Affiliation(s)
- Sumit Agarwal
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Michael Behring
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Hyung‐Gyoon Kim
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | | | | | - Nirzari Gupta
- Department of ChemistryUniversity of Alabama at BirminghamALUSA
| | - Prachi Bajpai
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | - Amr Elkholy
- Department of PathologyUniversity of Alabama at BirminghamALUSA
| | | | - Pran K. Datta
- Division of Hematology and OncologyDepartment of MedicineUniversity of Alabama at BirminghamALUSA
- Department of SurgeryUniversity of Alabama at BirminghamALUSA
- O'Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| | - Martin J. Heslin
- Department of SurgeryUniversity of Alabama at BirminghamALUSA
- O'Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| | - Sooryanarayana Varambally
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- O'Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| | - Upender Manne
- Department of PathologyUniversity of Alabama at BirminghamALUSA
- Department of SurgeryUniversity of Alabama at BirminghamALUSA
- O'Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamALUSA
| |
Collapse
|